US20220249486A1 - Setbp1 and xpo1 inhibitors for the treatment of sickle cell disease and beta-thalassemia - Google Patents

Setbp1 and xpo1 inhibitors for the treatment of sickle cell disease and beta-thalassemia Download PDF

Info

Publication number
US20220249486A1
US20220249486A1 US17/616,158 US202017616158A US2022249486A1 US 20220249486 A1 US20220249486 A1 US 20220249486A1 US 202017616158 A US202017616158 A US 202017616158A US 2022249486 A1 US2022249486 A1 US 2022249486A1
Authority
US
United States
Prior art keywords
compound
inhibitor
group
formula
setbp1
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US17/616,158
Inventor
Yang Du
Nhu NGUYEN
Kristbjorn Gudmundsson
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Henry M Jackson Foundation for Advancedment of Military Medicine Inc
Original Assignee
Henry M Jackson Foundation for Advancedment of Military Medicine Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Henry M Jackson Foundation for Advancedment of Military Medicine Inc filed Critical Henry M Jackson Foundation for Advancedment of Military Medicine Inc
Priority to US17/616,158 priority Critical patent/US20220249486A1/en
Publication of US20220249486A1 publication Critical patent/US20220249486A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41961,2,4-Triazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7105Natural ribonucleic acids, i.e. containing only riboses attached to adenine, guanine, cytosine or uracil and having 3'-5' phosphodiester links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/465Hydrolases (3) acting on ester bonds (3.1), e.g. lipases, ribonucleases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics

Definitions

  • the present disclosure relates to compounds and methods of inhibiting SETBP1 and XPO1 activities, enhancing the expression levels of embryonic and fetal hemoglobin molecules, and treating sickle cell disease and ⁇ -thalassemia.
  • ⁇ -globinopathies include sickle cell disease (SCD) and ⁇ -thalassemia and are among the most common monogenic disorders worldwide.
  • SCD results from the synthesis of an abnormal hemoglobin, which tends to aggregate under low oxygen conditions while ⁇ -thalassemia results from the reduced expression or absence of ⁇ -globins (Bauer et al., Blood 120(15): 2945-53 (2012)).
  • Large numbers of children are born worldwide with ⁇ -globinopathies each year, with approximately 300,000 having SCD and 40,000 having ⁇ -thalassemia (Weatherall, Blood 115(22): 4331-6 (2010)).
  • Bone marrow transplantation is currently the only curative treatment available for SCD in adults (Hsieh et al., N. Engl. J. Med. 361(24):2309-17 (2009); Hsieh et al., JAMA 312(1): 48-56 (2014)).
  • both the high cost associated with this procedure and the difficulty in finding matched donors have prevented its broad usage.
  • Fetal hemoglobin (HbF) expression can be induced by non-targeted drug treatments such as hydroxyurea (Esrick et al., Am. J. Hematol. 90(7): 624-8 (2015); Wong et al., Blood 124(26): 3850-7 (2014)).
  • hydroxyurea Error et al., Am. J. Hematol. 90(7): 624-8 (2015); Wong et al., Blood 124(26): 3850-7 (2014).
  • hydroxyurea the induction efficiency by hydroxyurea is low and treatment is not effective for all patients.
  • the present invention relates to unexpected findings that transcription factor SETBP1 regulates embryonic and fetal hemoglobin repression, and inhibition of SETBP1 can induce the expression of embryonic and fetal hemoglobins.
  • the present invention is directed to small molecule compounds that inhibit SETBP1 or XPO1 activities, and the use of these compounds in inducing the expression levels of embryonic and fetal hemoglobins that can be used as effective therapeutics for sickle cell disease and ⁇ -thalassemia.
  • a SETBP1 inhibitor for treating sickle cell disease in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a SETBP1 inhibitor, an XPO1 inhibitor, or a combination of a SETBP1 inhibitor and an XPO1 inhibitor.
  • FIG. 1 shows that Setbp1 knockdown increased embryonic and fetal hemoglobin expression levels in MEL cells.
  • Relative expression levels were calculated by normalizing to Rpl4 mRNA levels in the same sample and also to cells infected with shNC virus.
  • FIG. 2 shows that Compound (1AA) increased embryonic and fetal hemoglobin expression levels in MEL cells.
  • Relative expression levels were calculated by normalizing to Rpl4 mRNA levels in the same sample and also to cells infected with DMSO.
  • FIG. 3 shows that the XPO1 inhibitor KPT-185 increased embryonic and fetal hemoglobin expression levels in MEL cells.
  • KPT KPT-185
  • FIG. 4 shows the long-lasting effects of Compound (1AA) and KPT-185.
  • Real-time RT-PCR analysis of Hbb-bh1, Hbb-y, and Hbb-b1 mRNA levels in MEL cells 48 hours after removal of Compound (1AA) or KPT-185 from culture. The cells were first treated with Compound (1AA) or KPT-185 for 24 hours. Relative expression levels were calculated by normalizing to Rpl4 mRNA levels in the same sample and also to cells treated with DMSO.
  • FIG. 5 shows that KPT-185 is more effective than hydroxyurea in increasing embryonic hemoglobin expression in MEL cells.
  • FIG. 6 shows that Compound (1BB) increased embryonic and fetal hemoglobin expression levels in MEL cells.
  • FIG. 7 shows KPT-330 and KPT-8602 increased embryonic and fetal hemoglobin expression levels in MEL cells.
  • FIG. 8 shows that Compound 1AA and 1BB significantly increased embryonic and fetal hemoglobin expression levels in human erythroid cells.
  • Human CD34 + cells mobilized by granulocyte-colony stimulating factor (G-CSF) were induced to undergo erythroid differentiation in culture.
  • G-CSF granulocyte-colony stimulating factor
  • Treatment with Compound 1AA and 1BB at indicated concentrations or vehicles (DMSO or water) were started at day 5 of expansion condition.
  • Erythroid cells were harvested at day 10 of differentiation condition for real-time RT-PCR analysis of human embryonic s ( FIG. 8A ), fetal ⁇ ( FIG. 8B ), and adult ⁇ ( FIG. 8C ) hemoglobin mRNA levels.
  • FIG. 8D Representative bar graphs showing mRNA levels of human embryonic and fetal hemoglobin ( ⁇ + ⁇ ) as a percentage of total ⁇ -like globin ( ⁇ + ⁇ + ⁇ ) mRNA levels after treatment with Compound 1AA and 1BB versus DMSO. Note that a level of 20% or above for embryonic and fetal hemoglobin is considered curative for sickle cell disease. **, P ⁇ 0.01; ***, P ⁇ 0.001; ****P ⁇ 0.0001 (two-tailed Student's t test).
  • alkyl means any straight chain or branched, non-cyclic or cyclic, unsaturated or saturated aliphatic hydrocarbon containing, for example, from 1 to 20 carbon atoms, while the term “lower alkyl” has the same meaning as alkyl but contains from 1 to 5 carbon atoms.
  • the term “higher alkyl” has the same meaning as alkyl but contains from 6 to 20 carbon atoms.
  • saturated straight chain alkyls include, but are not limited to, methyl, ethyl, n-propyl, n-butyl, n-pentyl, n-hexyl, n-heptyl, n-octyl, n-nonyl, and the like; while saturated branched alkyls include, but are not limited to, isopropyl, sec-butyl, isobutyl, tert-butyl, isopentyl, and the like. Cyclic alkyls may be obtained by joining two alkyl groups bound to the same atom or by joining two alkyl groups each bound to adjoining atoms.
  • saturated cyclic alkyls include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like; while unsaturated cyclic alkyls include, but are not limited to, cyclopentenyl and cyclohexenyl, and the like.
  • Cyclic alkyls are also referred to herein as “cycloalkyls,” “homocycles” or “homocyclic rings.”
  • Unsaturated alkyls contain at least one double or triple bond between adjacent carbon atoms (referred to as an “alkenyl” or “alkynyl,” respectively).
  • Representative straight chain and branched alkenyls include, but are not limited to, ethylenyl, propylenyl, 1-butenyl, 2-butenyl, isobutylenyl, 1-pentenyl, 2-pentenyl, 3-methyl-1-butenyl, 2-methyl-2-butenyl, 2,3-dimethyl-2-butenyl, and the like; while representative straight chain and branched alkynyls include, but are not limited to, acetylenyl, propynyl, 1-butynyl, 2-butynyl, 1-pentynyl, 2-pentynyl, 3-methyl-1-butynyl, and the like.
  • aryl refers to any aromatic carbocyclic moiety containing, for example, 5 to 20 carbon atoms such as, but not limited to, phenyl or naphthyl.
  • arylalkyl refers to any alkyl having at least one alkyl hydrogen atom replaced with an aryl moiety, such as benzyl, but not limited to, (CH 2 ) 2 phenyl, —(CH 2 ) 3 phenyl, —CH(phenyl) 2 , and the like.
  • halogen refers to any fluoro, chloro, bromo, or iodo moiety.
  • haloalkyl refers to any alkyl having at least one hydrogen atom replaced with halogen, such as trifluoromethyl, and the like.
  • heteroaryl refers to any aromatic heterocycle ring of 5 to 20 members and having at least one heteroatom selected from nitrogen, oxygen and sulfur, and containing at least one carbon atom, including, but not limited to, both mono- and bicyclic ring systems.
  • heteroaryls include, but are not limited to, furyl, benzofuranyl, thiophenyl, benzothiophenyl, pyrrolyl, indolyl, isoindolyl, azaindolyl, pyridyl, quinolinyl, isoquinolinyl, oxazolyl, isooxazolyl, benzoxazolyl, pyrazolyl, imidazolyl, benzimidazolyl, thiazolyl, benzothiazolyl, isothiazolyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, cinnolinyl, phthalazinyl, or quinazolinyl.
  • heteroarylalkyl refers to any alkyl having at least one alkyl hydrogen atom replaced with a heteroaryl moiety, such as —CHpyridinyl, —CH 2 pyrimidinyl, and the like.
  • heterocycle refers to any non-aromatic 3- to 7-membered monocyclic or any non-aromatic 7- to 10-membered bicyclic heterocyclic ring which is either saturated or unsaturated and which contains from 1 to 4 heteroatoms independently selected from nitrogen, oxygen and sulfur, and wherein the nitrogen and sulfur heteroatoms may be optionally oxidized, and the nitrogen heteroatom may be optionally quaternized, including bicyclic rings in which any of the above heterocycles are fused to a benzene ring.
  • the heterocycle may be attached via any heteroatom or carbon atom.
  • Heterocycles may include, but are not limited to, morpholinyl, pyrrolidinonyl, pyrrolidinyl, piperidinyl, piperazinyl, imidazolinyl, imidazolidinyl, pyrazolinyl, pyrazolidinyl, 1,4-dioxanyl, dithianyl, hydantoinyl, valerolactamyl, oxiranyl, oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydropyridinyl, tetrahydroprimidinyl, tetrahydrothiophenyl, tetrahydrothiopyranyl, tetrahydropyrimidinyl, tetrahydrothiophenyl, tetrahydrothiopyranyl, and the like.
  • heterocycloalkyl refers to any alkyl having at least one alkyl hydrogen atom replaced with a heterocycle, such as —CH 2 morpholinyl, and the like.
  • cycloalkyl refers to any saturated or unsaturated (non-aromatic) carbocyclic ring containing from 3-7 carbon atoms, such as, but not limited to, cyclopropane, cyclobutane, cyclopentane, cyclohexane, cycloheptane, cyclohexene, and the like.
  • alkylamino refers to at least one alkyl moiety attached through a nitrogen bridge (i.e., —N-(alkyl)N, such as a dialkylamino) including, but not limited to, methylamino, ethylamino, dimethylamino, diethylamino, and the like.
  • alkyloxy refers to any alkyl moiety attached through an oxygen bridge (i.e., —O-alkyl) such as, but not limited to, methoxy, ethoxy, and the like.
  • alkylthio refers to any alkyl moiety attached through a sulfur bridge (i.e., —S— alkyl) such as, but not limited to, methylthio, ethylthio, and the like.
  • salts refers to any salt that complexes with identified compounds described herein.
  • examples of such salts include, but are not limited to, acid addition salts formed with inorganic acids (e.g., hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid, and the like), and salts formed with organic acids such as, but not limited to, acetic acid, oxalic acid, tartaric acid, succinic acid, malic acid, fumaric acid, maleic acid, ascorbic acid, benzoic acid, tannic acid, pamoic acid, alginic acid, polyglutamic, acid, naphthalene sulfonic acid, naphthalene disulfonic acid, and polygalacturonic acid.
  • inorganic acids e.g., hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid, and the like
  • organic acids such as, but not limited to, acetic acid,
  • Salt compounds can also be administered as pharmaceutically acceptable quaternary salts known to a person skilled in the art, which specifically includes the quaternary ammonium salts of the formula —NRR′R′′+Z—, wherein R, R′, R′′ is independently hydrogen, alkyl, or benzyl, and Z is a counter ion, including, but not limited to, chloride, bromide, iodide, alkoxide, toluenesulfonate, methylsulfonate, sulfonate, phosphate, or carboxylate (such as benzoate, succinate, acetate, glycolate, maleate, malate, fumarate, citrate, tartrate, ascorbate, cinnamoate, mandeloate, and diphenylacetate).
  • R, R′, R′′ is independently hydrogen, alkyl, or benzyl
  • Z is a counter ion, including, but not limited to, chloride, bromide, iodide
  • Salt compounds can also be administered as pharmaceutically acceptable pyridine cation salts having a substituted or unsubstituted partial formula: wherein Z is a counter ion, including, but not limited to, chloride, bromide, iodide, alkoxide, toluenesulfonate, methylsulfonate, sulfonate, phosphate, or carboxylate (such as benzoate, succinate, acetate, glycolate, maleate, malate, fumarate, citrate, tartrate, ascorbate, cinnamoate, mandeloate, and diphenylacetate).
  • Z is a counter ion, including, but not limited to, chloride, bromide, iodide, alkoxide, toluenesulfonate, methylsulfonate, sulfonate, phosphate, or carboxylate (such as benzoate, succinate, acetate, glycolate, maleate, malate, fum
  • pharmaceutically acceptable carrier includes any and all solvents, or a dispersion medium including, but not limited to, water, ethanol, a polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils, coatings, isotonic and absorption delaying agents, liposomes, commercially available cleansers, and the like. Supplementary bioactive ingredients also can be incorporated into such carriers.
  • the terms “reduce,” “inhibit,” “diminish,” “suppress,” “decrease,” “prevent” and grammatical equivalents when in reference to the expression of any symptom in an untreated subject relative to a treated subject, mean that the quantity and/or magnitude of the symptoms in the treated subject is lower than in the untreated subject by any amount that is recognized as clinically relevant by any medically trained personnel.
  • the quantity and/or magnitude of the symptoms in the treated subject is at least 10% lower than, at least 25% lower than, at least 50% lower than, at least 75% lower than, and/or at least 90% lower than the quantity and/or magnitude of the symptoms in the untreated subject.
  • induce when in reference to the expression of genes or proteins in a treated subject or a treated cell relative to a untreated subject or a untreated cell, mean that the quantity and/or levels of the genes or proteins in the treated subject or treated cell is greater than that in the untreated subject or untreated cell by any amount that is recognized as clinically relevant by any medically trained personnel.
  • the quantity and/or levels of the genes or proteins in the treated subject or treated cell is at least 10% greater than, at least 25% greater than, at least 50% greater than, at least 75% greater than, and/or at least 90% greater than the quantity and/or levels of the genes or proteins in the untreated subject or untreated cell.
  • inhibitory compound refers to any compound capable of interacting with (i.e., for example, attaching, binding etc.) to a binding partner under conditions such that the binding partner becomes unresponsive to its natural ligands.
  • Inhibitory compounds may include, but are not limited to, small organic molecules, antibodies, and proteins/peptides.
  • Attachment refers to any interaction between a medium (or carrier) and a drug. Attachment may be reversible or irreversible. Such attachment includes, but is not limited to, covalent bonding, ionic bonding, Van der Waals forces or friction, and the like.
  • a drug is attached to a medium (or carrier) if it is impregnated, incorporated, coated, in suspension with, in solution with, mixed with, etc.
  • drug refers to any pharmacologically active substance capable of being administered which achieves a desired effect.
  • Drugs or compounds can be synthetic or naturally occurring, non-peptide, proteins or peptides, oligonucleotides or nucleotides, polysaccharides or sugars.
  • administered refers to any method of providing a composition to a patient such that the composition has its intended effect on the patient.
  • An exemplary method of administering is by a direct mechanism such as, local tissue administration (i.e., for example, extravascular placement), oral ingestion, transdermal patch, topical, inhalation, suppository, etc.
  • patient is an animal, such as, for example, a mammal, such as, for example, a human that need not be hospitalized.
  • a mammal such as, for example, a human that need not be hospitalized.
  • out-patients and persons in nursing homes are “patients.”
  • a patient may comprise any age of a human or non-human animal and therefore includes both adult and juveniles (i.e., children). It is not intended that the term “patient” connote a need for medical treatment, therefore, a patient may voluntarily or involuntarily be part of experimentation whether clinical or in support of basic science studies.
  • subject refers to a vertebrate, preferably a mammal, more preferably a primate, still more preferably a human. Mammals include, without limitation, humans, primates, wild animals, feral animals, farm animals, sports animals and pets. The subject may be a juvenile or an adult.
  • XPO1 Exportin 1
  • CCM1 chromosomal maintenance 1
  • XPO1 inhibitor refers to an agent that inhibits or reduces the expression or biological activity of XPO1.
  • Example XPO1 inhibitors whether antibodies, inhibitory nucleic acids, or small molecules, are well-known in the art. Specific XPO1 inhibitors are identified in, e.g., Q. Sun et al., Signal Transduct Target Ther. 1:16010 (2016); U.S. Pat. No. 9,861,615; and K. Parikh et al., J. of Hematol. & Oncol 7:78 (2014).
  • SET binding protein 1 (“SETBP1”) is a protein that binds SET and is a DNA-binding protein known to regulate gene transcription.
  • a representative sequence of the SETBP1 protein is UniProt: Q9Y6X0 (or RefSeq: NP_001123582, NP_056374, NP_001366070, or NP_001366071).
  • the term “SETBP1 inhibitor” used herein refers to molecules that inhibit the activity of SETBP1 and/or inhibit the binding of SETBP1 to other proteins in a cell. Specific SETBP1 inhibitors, for example, are identified in WO2019/226773.
  • terapéuticaally effective amount or “effective amount” of a compound described herein or a pharmaceutically acceptable salt thereof means an amount sufficient to effect treatment when administered to a subject, to provide a therapeutic benefit such as amelioration of symptoms or slowing of disease progression.
  • a therapeutically effective amount may be an amount sufficient to decrease a symptom of a sickle cell disease.
  • the therapeutically effective amount may vary depending on the subject, and disease or condition being treated, the weight and age of the subject, the severity of the disease or condition, and the manner of administering, which can readily be determined by one of ordinary skill in the art.
  • SETBP1 is a repressor of the transcription of the embryonic and fetal hemoglobin genes.
  • small molecule SETBP1 inhibitors also demonstrated potent activation of embryonic and fetal hemoglobins.
  • An aspect of the invention is a method for treating a disease by activating the expression of embryonic hemoglobin in a subject, comprising administering to the subject a therapeutically effective amount of a SETBP1 inhibitor or an XPO1 inhibitor.
  • Another aspect of the invention is a method for treating a disease by activating expression of embryonic hemoglobin in a subject, comprising administering to a subject in need thereof a therapeutically effective amount of a SETBP1 inhibitor or a XPO1 inhibitor or a combination of a SETBP1 inhibitor and a XPO1 inhibitor.
  • Another aspect of the invention is a method for treating a disease by activating the expression of fetal hemoglobin in a subject, comprising administering to the subject a therapeutically effective amount of a SETBP1 inhibitor or an XPO1 inhibitor.
  • Another aspect of the invention is a method for treating a disease by activating expression of fetal hemoglobin in a subject, comprising administering to a subject in need thereof a therapeutically effective amount of a SETBP1 inhibitor or a XPO1 inhibitor or a combination of a SETBP1 inhibitor and a XPO1 inhibitor.
  • An exemplary embodiment of the invention is a method for treating sickle cell disease or ⁇ -thalassemia in a subject, comprising administering to the subject a therapeutically effective amount of a SETBP1 inhibitor.
  • Another exemplary embodiment of the invention is a method for treating sickle cell disease or D-thalassemia in a subject, comprising administering to the subject a therapeutically effective amount of an XPO1 inhibitor.
  • Another exemplary embodiment of the invention is a method for treating sickle cell disease or ⁇ -thalassemia in a subject, comprising administering to a subject in need thereof a therapeutically effective amount of a combination of a SETBP1 inhibitor and an XPO1 inhibitor.
  • Another exemplary embodiment of the invention is a method for treating sickle cell disease in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a SETBP1 inhibitor, an XPO1 inhibitor, or a combination of a SETBP1 inhibitor and an XPO1 inhibitor.
  • Another exemplary embodiment of the invention is a method for treating sickle cell disease (SCD) in a subject, comprising administering to a subject in need thereof a therapeutically effective amount of a SETBP1 inhibitor.
  • Another exemplary embodiment of the invention is a method for treating sickle cell disease (SCD) in a subject, comprising administering to a subject in need thereof a therapeutically effective amount of a XPO1 inhibitor.
  • Another exemplary embodiment of the invention is a method for treating sickle cell disease (SCD) in a subject, comprising administering to a subject in need thereof a therapeutically effective amount of a combination of a SETBP1 inhibitor and an XPO1 inhibitor.
  • sickle cell disease is treated with a SETBP1 inhibitor.
  • sickle cell disease is treated with an XPO1 inhibitor.
  • sickle cell disease is treated with a combination of SETBP1 inhibitor and an XPO1 inhibitor.
  • ⁇ -thalassemia is treated with a SETBP1 inhibitor.
  • ⁇ -thalassemia is treated with an XPO1 inhibitor.
  • ⁇ -thalassemia is treated with a combination of a SETBP1 inhibitor and an XPO1 inhibitor.
  • the SETBP1 inhibitor is an shRNA or an siRNA.
  • the shRNA or the siRNA has a sequence that is at least 95% identical to the nucleic acid sequences selected from the group consisting of SEQ ID NO: 1 and SEQ ID NO: 2.
  • the SETBP1 inhibitor is an anti-SETBP1 antibody inhibiting the activity of SETBP1 protein, a RNA molecule reducing SETBP1 transcription or translation, or a gene editing system disrupting the SETBP1 genomic sequence.
  • the XPO1 inhibitor is an shRNA or an siRNA.
  • the XPO1 inhibitor is an anti-XPO1 antibody inhibiting the activity of XPO1 protein, a RNA molecule reducing SETBP1 transcription or translation, or a gene editing system disrupting the XPO1 genomic sequence.
  • the SETBP1 inhibitor is a compound of Formula (1):
  • R 1 is —OR 8 , —SR 8 or —NR 8 R 8 ;
  • R 2 , R 3 , R 4 , R 5 and R 6 are independently H or C 1-4 alkyl
  • R 7 is —NR 10 R 10 or a heterocyclyl group
  • L is —(CR 9 R 9 ) n —;
  • X is an organic or inorganic anion
  • each R 8 is independently H or C 1-3 alkyl
  • each R 9 is independently H or C 1-3 alkyl
  • each R 10 is independently H or C 1-3 alkyl
  • n 1, 2, 3 or 4.
  • R 1 is —OR 8 .
  • R 1 is —OR 8 and R 3 is H.
  • R 1 is —OR 8
  • R 3 is H
  • R 8 is H
  • each R 9 is H.
  • n is 2.
  • R 1 is —OR 8 and at least one R 9 is H.
  • R 1 is —NR 8 R 8
  • R 3 is H
  • each R 8 is H
  • each R 9 is H.
  • At least one of R 4 and R 5 is —CH 3 .
  • each of R 4 and R 5 is —CH 3 .
  • R 1 is —OR 8
  • R 2 is H
  • R 3 is H
  • each of R 4 and R 8 is —CH 3 .
  • R 7 is a heterocyclyl group, where the heterocyclyl group is a 5- or 6-membered ring containing 1 or 2 nitrogen atoms.
  • R 7 is a heterocyclyl group
  • R 2 is H
  • R 1 is —OR 8 .
  • R 7 is a heterocyclyl group
  • R 2 is H
  • R 1 is —OR 8
  • R 3 is H.
  • R 7 is a heterocyclyl group where the hetereocyclyl group is a piperidinyl group, R 1 is —OR 8 , R 2 is H, R 3 is H, R 8 is H and each R 9 is H.
  • R 7 is a heterocyclyl group where the hetereocyclyl group is a piperidinyl group, R 2 is H and at least one R 9 is H.
  • R 7 is a heterocyclyl group where the hetereocyclyl group is a piperidinyl group, R 2 is H and each R 9 is H.
  • R 7 is a heterocyclyl group where the hetereocyclyl group is a piperidinyl group, R 2 is H and at least one of R 4 and R 8 is —CH 3 .
  • R 7 is a heterocyclyl group where the hetereocyclyl group is a piperidinyl group, R 2 is H and each of R 4 and R 5 is —CH 3 .
  • R 7 is a heterocyclyl group where the hetereocyclyl group is a piperidinyl group, R 1 is —OR 8 , R 2 is H, R 3 is H and each of R 4 and R 5 is —CH 3 .
  • X is F, Cl, Br or I.
  • R 1 is a heterocyclyl group where the hetereocyclyl group is a piperidinyl group, R 1 is —OR 8 , R 2 is H, R 3 is H, each of R 4 and R 5 is —CH 3 and X is F, Cl, Br or I.
  • R 7 is —NR 10 R 10 .
  • R 7 is —NR 10 R 10 and each R 10 is independently C 1-3 alkyl.
  • R 7 is —NR 10 R 10 and one R 10 is C 1-3 alkyl and one R 10 is —H.
  • R 2 is H
  • R 7 is —NR 10 R 10 and each R 10 is —CH 2 CH 3 .
  • R 2 is H
  • R 7 is —NR 10 R 10 and each R 10 is H.
  • R 2 is H
  • R 7 is —NR 10 R 10
  • R 1 is —OR 8
  • R 3 is H.
  • R 2 is H
  • R 7 is —NR 10 R 10
  • R 1 is —OR 8
  • R 3 is H
  • R 8 is H and each R 9 is H.
  • R 2 is H
  • R 7 is —NR 10 R 10 and at least one R 9 is H.
  • R 2 is H
  • R 7 is —NR 10 R 10 and each R 9 is H.
  • R 7 is —NR 10 R 10 , each R 10 is —CH 2 CH 3 and at least one of R 4 and R 5 is —CH 3 .
  • R 2 is H
  • R 7 is —NR 10 R 10
  • each R 10 is —CH 2 CH 3
  • each of R 4 and R 5 is —CH 3 .
  • R 2 is H
  • R 7 is —NR 10 R 10
  • each R 10 is —CH 2 CH 3
  • R 1 is —OR 8
  • R 3 is H
  • each of R 4 and R 5 is —CH 3
  • X is F, Cl, Br or I.
  • R 7 is a heterocyclyl group selected from the group consisting of morpholinyl, pyrrolidinonyl, pyrrolidinyl, piperidinyl, piperazinyl and tetrahydropyranyl.
  • R 7 is a heterocyclyl group selected from the group consisting of morpholinyl, piperidinyl, piperazinyl and tetrahydropyranyl.
  • R 1 is —OR 8 ;
  • R 2 , R 3 and R 6 are H;
  • R 4 and R 5 are independently C 1-3 alkyl;
  • R 7 is —NR 10 R 10 or a piperidinyl group;
  • L is —(CR 9 R 9 ) n —;
  • X is an organic or inorganic anion;
  • X is selected from the group consisting of F, Cl, Br and I.
  • n is 2.
  • R 1 is —OR 8 ;
  • R 2 , R 3 , R 6 , R 8 and R 9 are H;
  • R 4 and R 5 are —CH 3 ;
  • R 7 is a piperidinyl group;
  • L is —(CR 9 R 9 ) n —;
  • X is selected from the group consisting of F, Cl, Br and I; and
  • n is 1, 2, 3 or 4.
  • R 1 is —OR 8 ;
  • R 2 , R 3 , R 6 , R 8 and R 9 are H;
  • R 4 and R 5 are —CH 3 ;
  • R 7 is —NR 10 R 10 ;
  • each R 10 is independently C 1-3 alkyl;
  • L is —(CR 9 R 9 ) n —;
  • X is selected from the group consisting of F, Cl, Br and I; and
  • n is 1, 2, 3 or 4.
  • the compound of Formula (1) is a compound of Formula (1A):
  • X is an organic or inorganic anion.
  • X is selected from the group consisting of halo (F, Cl, Br, I), alkoxide, p-toluenesulfonate, methylsulfonate, sulfonate, phosphate, and carboxylate (such as benzoate, succinate, acetate, glycolate, maleate, malate, fumarate, citrate, tartrate, ascorbate, cinnamoate, mandeloate, and diphenylacetate).
  • X is selected from the group consisting of F, Cl, Br, and I.
  • the compound of Formula (1) is Compound (1AA):
  • the compound of Formula (1) is a compound of Formula (1B):
  • X is an organic or inorganic anion.
  • X is selected from the group consisting of halo (F, Cl, Br, I), alkoxide, p-toluenesulfonate, methylsulfonate, sulfonate, phosphate, and carboxylate (such as benzoate, succinate, acetate, glycolate, maleate, malate, fumarate, citrate, tartrate, ascorbate, cinnamoate, mandeloate, and diphenylacetate).
  • X is selected from the group consisting of F, Cl, Br and I.
  • the compound of Formula (1) is Compound (1BB):
  • the SETBP1 inhibitor is a compound of Formula (2):
  • R 1 is an aryl group or a heteroaryl group, where the aryl group and the heteroaryl group each has 0 to 3 substituents independently selected from the group consisting of —NR 6 R 6 , OH, F, Cl, Br and C 1-3 alkyl;
  • R 2 and R 3 are independently H or C 1-6 alkyl
  • R 4 and R 5 are independently H, —NR 6 R 6 or OR 6 ;
  • each R 6 is independently H or C 1-3 alkyl
  • X 1 and X2 are independently CH or N;
  • L is —(CR 7 R 7 ) n —;
  • each R 7 is independently H or C 1-3 alkyl
  • n 1, 2, 3 or 4.
  • the aryl or heteroaryl group of R 1 is selected from the group consisting of phenyl, furyl, benzofuranyl, thiophenyl, benzothiophenyl, pyrrolyl, pyridyl and pyrimidinyl.
  • R 1 is a phenyl group having 1 substituent that is —NR 6 R 6 .
  • R 1 is a phenyl group having 1 substituent that is —NR 6 R 6 where at least one of R 6 is H.
  • R 1 is a phenyl group having 1 substituent that is —NR 6 R 6 where each R 6 is H.
  • At least one of R 2 and R 3 is H.
  • each of R 2 and R 3 is H.
  • X 1 and X2 are each N and at least one of R 4 and R 5 is —NR 6 R 6 .
  • X 1 and X2 are each N and each of R 4 and R 5 is —NR 6 R 6 .
  • X 1 and X2 are each N and at least one of R 4 and R 5 is —NR 6 R 6 where each R 6 is H.
  • X 1 and X2 are each N and each of R 4 and R 5 is —NR 6 R 6 where each R 6 is H.
  • L is —(CR 7 R 7 )— and each R 7 is independently H or C 1-3 alkyl. In an exemplary embodiment of Formula (2), L is —CH 2 —.
  • the compound of Formula (2) is a compound of Formula (2A):
  • HA is selected from the group consisting of inorganic acids (e.g., hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid, and the like) and organic acids (e.g., acetic acid, oxalic acid, tartaric acid, succinic acid, malic acid, fumaric acid, maleic acid, ascorbic acid, benzoic acid, tannic acid, pamoic acid, alginic acid, polyglutamic, acid, naphthalene sulfonic acid, naphthalene disulfonic acid and polygalacturonic acid).
  • HA is selected from the group consisting of hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid and nitric acid.
  • the compound of Formula (2) is Compound (2AA):
  • the compound of Formula (1), Formula (1A), Compound (1AA), Formula (1B), or Compound (1BB) is present in a pharmaceutical composition comprising one or more excipients.
  • the compound of Formula (1) is present in a pharmaceutical composition comprising one or more excipients.
  • the compound of Formula (1A) is present in a pharmaceutical composition comprising one or more excipients.
  • Compound (1AA) is present in a pharmaceutical composition comprising one or more excipients.
  • the compound of Formula (1B) is present in a pharmaceutical composition comprising one or more excipients.
  • Compound (1BB) is present in a pharmaceutical composition comprising one or more excipients.
  • the compound of Formula (2), Formula (2A), or Compound (2AA) is present in a pharmaceutical composition comprising one or more excipients.
  • the compound of Formula (2) is present in a pharmaceutical composition comprising one or more excipients.
  • the compound of Formula (2A) is present in a pharmaceutical composition comprising one or more excipients.
  • Compound (2AA) is present in a pharmaceutical composition comprising one or more excipients.
  • a SETBP1 inhibitor and an XPO1 inhibitor are present in a pharmaceutical composition comprising one or more excipients.
  • the SETBP1 inhibitor or the XPO1 inhibitor or the combination of the SETBP1 inhibitor and the XPO1 inhibitor is present in a pharmaceutical composition comprising one or more excipients.
  • the XPO1 inhibitor is selected from the group consisting of KPT-185, KPT-276, KPT-330, KPT-335, KPT-8602, and combination thereof. In an exemplary embodiment, the XPO1 inhibitor is selected from the group consisting of KPT-185, KPT-276, KPT-330, KPT-335 and KPT-8602.
  • An exemplary embodiment of the invention is a method for treating sickle cell disease (SCD) or ⁇ -thalassemia in a subject, comprising administering to the subject a therapeutically effective amount of a compound of Formula (1) (such as a compound of Formula (1A), such as Compound (1AA), such as a compound of Formula (1), such as Compound (1 TB)) or a compound of Formula (2) (such as a compound of Formula (2A), such as Compound (2AA)).
  • a compound of Formula (1) such as a compound of Formula (1A), such as Compound (1AA), such as a compound of Formula (1), such as Compound (1 TB)
  • a compound of Formula (2) such as a compound of Formula (2A), such as Compound (2AA)
  • An exemplary embodiment of the invention is a method for treating sickle cell disease (SCD) or ⁇ -thalassemia in a subject, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of Formula (1) (such as a compound of Formula (1A), such as Compound (1AA), or such as a compound of Formula (1B), such as Compound (1B)).
  • a compound of Formula (1) such as a compound of Formula (1A), such as Compound (1AA), or such as a compound of Formula (1B), such as Compound (1B)
  • An exemplary embodiment of the invention is a method for treating sickle cell disease (SCD) or ⁇ -thalassemia in a subject, comprising administering to a subject in need thereof a therapeutically effective amount of a combination of a compound of Formula (1) (such as a compound of Formula (1A), such as Compound (1AA), or such as a compound of Formula (1B), such as Compound (1B)) and a XPO1 inhibitor.
  • a compound of Formula (1) such as a compound of Formula (1A), such as Compound (1AA), or such as a compound of Formula (1B), such as Compound (1B)
  • An exemplary embodiment of the invention is a method for treating sickle cell disease (SCD) or ⁇ -thalassemia in a subject, comprising administering to a subject in need thereof a therapeutically effective amount of a combination of a compound of Formula (1) (such as a compound of Formula (1A), such as Compound (1AA), or such as a compound of Formula (1B), such as Compound (1B)) and a XPO1 inhibitor selected from the group consisting of KPT-185, KPT-276, KPT-330, KPT-335, KPT-8602 and combinations thereof.
  • a compound of Formula (1) such as a compound of Formula (1A), such as Compound (1AA), or such as a compound of Formula (1B), such as Compound (1B)
  • a XPO1 inhibitor selected from the group consisting of KPT-185, KPT-276, KPT-330, KPT-335, KPT-8602 and combinations thereof.
  • An exemplary embodiment of the invention is a method for treating sickle cell disease or ⁇ -thalassemia in a subject, comprising administering to the subject a therapeutically effective amount of a combination of a compound of Formula (1) (such as a compound of Formula (1A), such as Compound (1AA), or such as a compound of Formula (1B), such as Compound (1BB)) or a compound of Formula (2) (such as a compound of Formula (2A), such as Compound (2AA)) and an XPO1 inhibitor selected from the group consisting of KPT-185, KPT-276, KPT-330, KPT-335, KPT-8602 and combinations thereof.
  • a compound of Formula (1) such as a compound of Formula (1A), such as Compound (1AA), or such as a compound of Formula (1B), such as Compound (1BB)
  • a compound of Formula (2) such as a compound of Formula (2A), such as Compound (2AA)
  • an XPO1 inhibitor selected from the group consisting of KPT-185, KPT-276, K
  • Another exemplary embodiment of the invention is a method for treating sickle cell disease or ⁇ -thalassemia in a subject, comprising administering to the subject a therapeutically effective amount of a XPO1 inhibitor selected from the group consisting of KPT-185, KPT-276, KPT-330, KPT-335, KPT-8602 and combinations thereof.
  • a XPO1 inhibitor selected from the group consisting of KPT-185, KPT-276, KPT-330, KPT-335, KPT-8602 and combinations thereof.
  • Another exemplary embodiment of the invention is a method for treating sickle cell disease (SCD) in a subject, comprising administering to a subject in need thereof a therapeutically effective amount of a XPO1 inhibitor selected from the group consisting of KPT-185, KPT-276, KPT-330, KPT-335, KPT-8602 and combinations thereof.
  • a XPO1 inhibitor selected from the group consisting of KPT-185, KPT-276, KPT-330, KPT-335, KPT-8602 and combinations thereof.
  • Another exemplary embodiment of the invention is a method for treating ⁇ -thalassemia in a subject, comprising administering to the subject a therapeutically effective amount of a SETBP1 inhibitor or a combination of a SETBP1 inhibitor and an XPO1 inhibitor.
  • Another exemplary embodiment of the invention is a method for treating 3-thalassemia in a subject, comprising administering to the subject a therapeutically effective amount of a SETBP1 inhibitor.
  • Another exemplary embodiment of the invention is a method for treating ⁇ -thalassemia in a subject, comprising administering to the subject a therapeutically effective amount of a combination of a SETBP1 inhibitor and an XPO1 inhibitor.
  • the subject is an adult human subject or a juvenile human subject.
  • the human subject is an adult human subject.
  • the human subject is a juvenile human subject.
  • Some embodiments provides for a method of inducing embryonic and fetal hemoglobin expression in a cell, wherein the cell is a hematopoietic progenitor cell, an erythroid progenitor cell, or an erythroid cell, comprising contacting the cell with a SETBP1 inhibitor, an XPO1 inhibitor, or a combination of a SETBP1 inhibitor and an XPO1 inhibitor.
  • the invention provides a pharmaceutical composition comprising at least one pharmaceutically acceptable carrier, in addition to one or more of the compounds described herein.
  • the composition can be present in any suitable form for the desired route of administration.
  • any suitable orally deliverable dosage form can be used, including, without limitation, tablets, capsules (solid or liquid filled), powders, granules, syrups and other liquids, elixirs, inhalants, troches, lozenges and solutions.
  • Injectable compositions or i.v. infusions are also provided in the form of solutions, suspensions, and emulsions.
  • the compounds of the invention can be formulated as described herein and are suitable for administration in a therapeutically effective amount to the subject in any number of ways.
  • a therapeutically effective amount of a compound as described herein depends upon the amounts and types of excipients employed, the amounts and specific types of active ingredients present in a dosage form, and the route by which the compound is to be administered to patients.
  • Typical dosage levels for the compounds of the invention generally range from about 0.001 to about 100 mg per kg of the subject's body weight per day which can be administered in single or multiple doses.
  • An exemplary dosage is about 0.01 to about 25 mg/kg per day or about 0.05 to about 10 mg/kg per day.
  • the dosage level ranges from about 0.01 to about 25 mg/kg per day, such as about 0.05 to about 10 mg/kg per day, or about 0.1 to about 5 mg/kg per day.
  • a dose can typically range from about 0.1 mg to about 2000 mg per day and can be given as a single once-a-day dose or, alternatively, as divided doses throughout the day, optionally taken with food.
  • the daily dose is administered twice daily in equally divided doses.
  • a daily dose range can range from about 5 mg to about 500 mg per day such as, for example, between about 10 mg and about 300 mg per day.
  • the therapy can be initiated at a lower dose, such as from about 1 mg to about 25 mg, and increased if necessary up to from about 200 mg to about 2000 mg per day as either a single dose or divided doses, depending on the subject's global response.
  • Suitable oral compositions in accordance with the invention include, without limitation, tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, syrups or elixirs.
  • Inventive compositions suitable for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions.
  • liquid formulations of the compounds can contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations of the active agents.
  • typical non-toxic pharmaceutically acceptable excipients include, without limitation, inert diluents such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents such as, for example, corn starch, or alginic acid; binding agents such as, for example, starch, gelatin or lubricating agents such as, for example, magnesium stearate, stearic acid or talc.
  • the tablets may be uncoated or, alternatively, they may be coated by known coating techniques to delay disintegration and absorption in the gastrointestinal tract and thereby to provide a sustained therapeutic action over a desired time period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate may be employed.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent such as, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium such as, for example peanut oil, liquid paraffin or olive oil.
  • an inert solid diluent such as, for example, calcium carbonate, calcium phosphate or kaolin
  • water or an oil medium such as, for example peanut oil, liquid paraffin or olive oil.
  • the compound is admixed with excipients suitable for maintaining a stable suspension.
  • excipients include, without limitation, sodium carboxymethylcellulose, methylcellulose, hydropropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia.
  • Oral suspensions can also contain dispersing or wetting agents, such as naturally-occurring phosphatide such as, for example, lecithin, or condensation products of an alkylene oxide with fatty acids such as, for example, polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols such as, for example, heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as, for example, polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides such as, for example, polyethylene sorbitan monooleate.
  • dispersing or wetting agents such as naturally-occurring phosphatide such as, for example, lecithin, or condensation products of an alkylene oxide with fatty acids such as, for example, polyoxyethylene stearate, or condensation products of ethylene oxide with long chain
  • the aqueous suspensions may also contain one or more preservatives such as, for example, ethyl or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents such as sucrose or saccharin.
  • preservatives such as, for example, ethyl or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents such as sucrose or saccharin.
  • Sweetening agents such as those set forth above, and flavoring agents may be added to provide palatable oral preparations. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water can provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
  • a dispersing or wetting agent e.g., talc, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, mannitol, mannitol, mannitol, mannitol, mannitol, mannitol, mannitol, mannitol
  • Syrups and elixirs may be formulated with sweetening agents such as, for example, glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, and flavoring and coloring agents.
  • sweetening agents such as, for example, glycerol, propylene glycol, sorbitol or sucrose.
  • Such formulations may also contain a demulcent, a preservative, and flavoring and coloring agents.
  • compositions for parenteral administrations are formulated in a sterile medium suitable for intravenous, intramuscular or intrathecal delivery.
  • a sterile injectable preparation of the compounds may be in the form of a sterile injectable solution or sterile injectable suspension.
  • Non-toxic, parentally acceptable diluents or solvents such as, for example, 1,3-butanediol can be used to formulate the parenteral compositions.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile oils also can be employed as a solvent or a suspending medium.
  • any bland fixed oil may be employed, including synthetic monoglycerides or diglycerides.
  • fatty acids such as oleic acid can be used in the preparation of injectables.
  • the parenteral formulation can contain other adjuvants such as local anesthetics, preservatives and buffering agents.
  • Gene editing technologies may include, but are not limited to, a CRISPR/Cas9 system, a CRISPR/Cas13 system, a zinc finger nuclease system, a TALEN system, and a meganuclease system. These gene editing technologies themselves are described in public domain (e.g. U.S. Pat. No. 10,196,652, Science: Vol. 358, Issue 6366, pp. 1019-1027. DOI: 10.1126/science.aaq0180).
  • the SETBP1 inhibitor is one or more components of a gene editing system targeting one or more sites within a gene encoding SETBP1 or a regulatory element thereof, a nucleic acid molecule encoding the one or more components of the gene editing system, or a combination thereof.
  • the SETBP1 inhibitor is a gene editing system, and wherein the gene editing system is selected from the group consisting of CRISPR/Cas9, CRISPR/Cas13, a zinc finger nuclease system, a TALEN system, and a meganuclease system.
  • the gene editing system is a CRISPR/Cas9 system.
  • the gene editing system is a CRISPR/Cas13 system. In some embodiments, the gene editing system is a zinc finger nuclease system. In some embodiments, the gene editing system is a TALEN system. In some embodiments, the gene editing system is a meganuclease system.
  • the SETBP1 inhibitor comprises a guide RNA molecule comprising a tracr and a crRNA. In some embodiments, the crRNA comprises a targeting domain that is complementary with a target sequence of SETBP1.
  • the XPO1 inhibitor is one or more components of a gene editing system targeting one or more sites within a gene encoding XPO1 or a regulatory element thereof, a nucleic acid molecule encoding the one or more components of the gene editing system, or a combination thereof.
  • the XPO1 inhibitor is a gene editing system, and wherein the gene editing system is selected from the group consisting of CRISPR/Cas9, CRISPR/Cas13, a zinc finger nuclease system, a TALEN system, and a meganuclease system.
  • the gene editing system is a CRISPR/Cas9 system.
  • the gene editing system is a CRISPR/Cas13 system. In some embodiments, the gene editing system is a zinc finger nuclease system. In some embodiments, the gene editing system is a TALEN system. In some embodiments, the gene editing system is a meganuclease system.
  • the XPO1 inhibitor comprises a guide RNA molecule comprising a tracr and a crRNA. In some embodiments, the crRNA comprises a targeting domain that is complementary with a target sequence of XPO1.
  • compositions according to the invention may contain one or more additional therapeutic agents, for example, to increase efficacy and/or to decrease side effects.
  • additional therapeutic agents include, without limitation, agents to treat or inhibit immunological, inflammatory, autoimmune or allergic disorders.
  • the cells were treated with selected inhibitors or vehicle in a culture medium (RPM 11640 medium with 10% fetal bovine serum) for 48 hours and were subsequently harvested for analysis of relative mRNA levels of Hbb-bh1, Hbb-y, and Hbb-b1 using real-time RT-PCR.
  • a culture medium RPM 11640 medium with 10% fetal bovine serum
  • the cells were treated with inhibitors or vehicle following or concurrent with induction of erythroid differentiation using an established two-phase culture protocol (Xu et al., Genes & Development 24(8): 783-798 (2010)). Relative mRNA levels of ⁇ -like globin genes in human cells after treatment were also analyzed using real-time RT-PCR.
  • the non-targeting control pLKO.1 lentiviral shRNA construct shNC was purchased from Sigma (SHC002).
  • Lentiviral shRNA constructs targeting Setbp1 (sh84 and sh8) were generated by cloning Setbp1 targeting sequences (sh84, CGG CTT TGA ATC CCA ATC ATT (SEQ ID NO:1); sh8, CCT ATG ATG AAC CTT GGT TAT (SEQ ID NO:2)) into pLKO.1.
  • the constructs were co-transfected using FuGENE 6 transfection reagent into 293T cells along with packaging plasmid A8.9 and a plasmid expressing VSV-G, and virus were harvested at 72 hours after transfection.
  • Viral titers were calculated by infecting NIH-3T3 cells with serial dilutions of viral stocks and counting puromycin resistant colonies. Lentiviral infections were performed by spinoculation in which a mixture of lentivirus and MEL cells at 4:1 ratio in 24-well plates were spun at 2000 ⁇ g for 90 minutes at 37° C. Puromycin (2 ⁇ g/mL) was added to culture at 24 hours after spinoculation to select for infected cells.
  • Setbp1 plays a role in the repression of embryonic and fetal hemoglobin expression
  • the expression levels of Setbp1 were knocked down using two different lentiviral shRNAs in MEL cells, which is a mouse erythroid progenitor cell line conventionally used for studying repression of embryonic and fetal hemoglobins.
  • the Setbp1 knockdowns significantly increased the mRNA levels of embryonic (Hbb-bh1) and fetal (Hbb-y) hemoglobin genes ( FIG. 1 ), suggesting that Setbp1 is a repressor of their transcription.
  • Compound (1AA) which has been identified as an inhibitor of SETBP1 transcriptional activity (WO2019/226773), was tested for its effect on the expression of Hbb-bh1 and Hbb-v in MEL cells. Consistent with Setbp1 knockdown studies, treatment with Compound (1AA) at 0.1 ⁇ M for 48 hours led to over an 8-fold increase in the mRNA levels of both Hbb-bh1 and Hbb-y ( FIG. 2 ). This unexpectedly potent activating effect of Compound (1AA) appears limited to embryonic and fetal hemoglobin since only minor changes in the levels of adult hemoglobin Hbb-b1 were detected under the same conditions ( FIG. 2 ).
  • Compound (1BB) another inhibitor of SETBP1 (WO2019/226773), was also observed to significantly increase the expression of Hbb-bh1 and Hbb-y in MEL cells while having minor effects on Hbb-b1 mRNA levels ( FIG. 6 ).
  • Compound (1AA) has been shown to inhibit the activity of SETBP1 by blocking its interaction with key cofactor XPO1 (WO2019/226773).
  • This interaction between SETBP1 and XPO1 can also be blocked by XPO1 inhibitors such as, but not limited to, KPT-185 and KPT-330. Therefore, XPO1 inhibitors were also tested for their effect on the expression of Hbb-bh1 and Hbb-y in MEL cells.
  • Treatment with 0.1 ⁇ m KPT-185 for 48 hours was observed to increase mRNA levels of Hbb-bh1 and Hbb-y by over 60-fold and over 14-fold, respectively ( FIG. 3 ).
  • XPO1 inhibitors KPT-330 and KPT-8602 also were found to significantly increase mRNA levels of Hbb-bh1 and Hbb-y in MEL cells ( FIG. 7 ). These results clearly show that XPO1 inhibitors are also surprisingly efficient in activating embryonic and fetal hemoglobin expression. Similar to Compounds (1AA) and (1BB), XPO1 inhibitors also a exerted minimal effect on the expression levels of Hbb-b1 ( FIG. 2 and FIG. 7 ).
  • a further experiment examined whether continuous treatment of Compound (1AA) and KPT-185 as exemplary SETBP1 and XPO1 inhibitors, respectively, was required for activation of Hbb-bh1 and Hbb-y expression.
  • MEL cells were treated with Compound (1AA) and KPT-185 for 24 hours and the Hbb-bh1 and Hbb-y mRNA levels were then examined at 48 hours after their removal.
  • Significantly elevated levels of Hbb-bh1 and Hbb-y mRNA were still detected in MEL cells ( FIG. 4 ).
  • KPT-185 The ability of KPT-185 to activate Hbb-bh1 and Hbb-y expression in MEL cells was also compared to that of hydroxyurea, an FDA-approved drug for sickle cell disease. While KPT-185 was observed to activate Hbb-y expression to similar levels to that of hydroxyurea, surprisingly KPT-185 was substantially more effective than hydroxyurea in activating Hbb-bh1 expression with almost 30-fold higher levels of Hbb-bh1 mRNA being detected in cells treated with KPT-185 compared to cells treated with hydroxyurea ( FIG. 5 ). This finding shows that XPO1 inhibitors such as KPT-185 are more effective than hydroxyurea in treating sickle cell disease.
  • cells were transferred into differentiation medium containing StemSpan SFEM Medium with 2% Penicillin-Streptomycin, 20 ng/ml SCF (Biolegend), 1 U/ml EPO (Amgen), 5 ng/ml IL-3 (Biolegend), 2 ⁇ M dexamethasone (Sigma), and 1 ⁇ M ⁇ -estradiol (Sigma).
  • Cells were maintained at a density of 0.1-1 ⁇ 10 6 cells/mL in differentiation medium for 10 days with media changes every two days. Drug treatments were started at day 5 of expansion condition and maintained thereafter. Fresh drugs were added at each medium change. The data is summarized in FIG. 8 .

Abstract

The present disclosure relates to compounds that inhibit SETBP1 or XP01 activities. Also disclosed are methods of using such compounds to increase the expression of embryonic and fetal hemoglobin molecules, and to treat sickle cell disease and β-thalassemia. The present invention relates to unexpected findings that transcription factor SETBP1 regulates embryonic and fetal hemoglobin repression, and inhibition of SETBP1 can induce the expression of embryonic and fetal hemoglobins.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims priority to U.S. Provisional Application No. 62/858,103, filed Jun. 6, 2019, which is incorporated herein by reference for all purposes.
  • STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
  • This disclosure was made with government support under grant numbers HU00011920059 and PED-86-4142 awarded by the Uniformed Services University of the Health Sciences (USUHS). The government has certain rights in the invention.
  • SEQUENCE LISTING
  • The instant application contains a Sequence Listing which has been submitted in ASCII format via EFS-Web and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Jun. 4, 2020, is named 316990_ST25.txt and is 702 bytes in size.
  • FIELD
  • The present disclosure relates to compounds and methods of inhibiting SETBP1 and XPO1 activities, enhancing the expression levels of embryonic and fetal hemoglobin molecules, and treating sickle cell disease and β-thalassemia.
  • BACKGROUND
  • β-globinopathies include sickle cell disease (SCD) and β-thalassemia and are among the most common monogenic disorders worldwide. SCD results from the synthesis of an abnormal hemoglobin, which tends to aggregate under low oxygen conditions while β-thalassemia results from the reduced expression or absence of β-globins (Bauer et al., Blood 120(15): 2945-53 (2012)). Large numbers of children are born worldwide with β-globinopathies each year, with approximately 300,000 having SCD and 40,000 having β-thalassemia (Weatherall, Blood 115(22): 4331-6 (2010)).
  • Treatment and management of SCD is mostly focused on pain relief and blood transfusions (Yawn et al., JAMA 312(10): 1033-48 (2014)). In β-thalassemia, blood transfusions are often needed throughout the patient's life (Olivieri et al., Cold Spring Harb. Perspect. Med. 3(6) (2013)). However, repeated blood transfusions can lead to iron overload, which is a major complication affecting both cardiovascular and liver functions. As a result, other therapeutic options are being actively explored, including bone marrow transplantation and treatments to reactivate the expression of embryonic and fetal hemoglobins (Bauer et al., Blood 120(15): 2945-53 (2012)). Bone marrow transplantation is currently the only curative treatment available for SCD in adults (Hsieh et al., N. Engl. J. Med. 361(24):2309-17 (2009); Hsieh et al., JAMA 312(1): 48-56 (2014)). However, both the high cost associated with this procedure and the difficulty in finding matched donors have prevented its broad usage.
  • Fetal hemoglobin (HbF) expression can be induced by non-targeted drug treatments such as hydroxyurea (Esrick et al., Am. J. Hematol. 90(7): 624-8 (2015); Wong et al., Blood 124(26): 3850-7 (2014)). However, the induction efficiency by hydroxyurea is low and treatment is not effective for all patients. Effective small molecules for inducing embryonic and fetal hemoglobin expression, and thereby treating SCD and β-thalassemia, remain to be identified.
  • SUMMARY
  • The present invention relates to unexpected findings that transcription factor SETBP1 regulates embryonic and fetal hemoglobin repression, and inhibition of SETBP1 can induce the expression of embryonic and fetal hemoglobins.
  • In some embodiments, the present invention is directed to small molecule compounds that inhibit SETBP1 or XPO1 activities, and the use of these compounds in inducing the expression levels of embryonic and fetal hemoglobins that can be used as effective therapeutics for sickle cell disease and β-thalassemia.
  • Provided herein are methods for treating sickle cell disease in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a SETBP1 inhibitor, an XPO1 inhibitor, or a combination of a SETBP1 inhibitor and an XPO1 inhibitor.
  • Also provided herein are methods for treating β-thalassemia in a subject, comprising administering to the subject a therapeutically effective amount of a SETBP1 inhibitor or a combination of a SETBP1 inhibitor and an XPO1 inhibitor.
  • Also provided herein are methods of inducing embryonic and fetal hemoglobin expression in a cell, wherein the cell is a hematopoietic progenitor cell, an erythroid progenitor cell, or an erythroid cell, comprising contacting the cell with a SETBP1 inhibitor, an XPO1 inhibitor, or a combination of a SETBP1 inhibitor and an XPO1 inhibitor.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The following figures are merely illustrative of specific embodiments of the invention and are not intended to otherwise limit the full scope of the invention as described.
  • FIG. 1 shows that Setbp1 knockdown increased embryonic and fetal hemoglobin expression levels in MEL cells. Real-time RT-PCR analysis of Hbb-bh1 and Hbb-y mRNA levels in MEL cells at 72 hours after infection with lentiviral shRNAs targeting Setbp1 (sh84 and sh8) or non-targeting control lentiviral shRNA (shNC). Relative expression levels were calculated by normalizing to Rpl4 mRNA levels in the same sample and also to cells infected with shNC virus.
  • FIG. 2 shows that Compound (1AA) increased embryonic and fetal hemoglobin expression levels in MEL cells. Real-time RT-PCR analysis of Hbb-bh1, Hbb-y, and Hbb-b1 mRNA levels in MEL cells after 48 hours of treatment with compound 1AA at indicated concentration or with control DMSO. Relative expression levels were calculated by normalizing to Rpl4 mRNA levels in the same sample and also to cells infected with DMSO.
  • FIG. 3 shows that the XPO1 inhibitor KPT-185 increased embryonic and fetal hemoglobin expression levels in MEL cells. Real-time RT-PCR analysis of Hbb-bh1, Hbb-y, and Hbb-b1 mRNA levels in MEL cells after 48 hours of treatment with KPT-185 (KPT) at indicated concentration or with control DMSO. Relative expression levels were calculated by normalizing to Rpl4 mRNA levels in the same sample and also to cells infected with DMSO.
  • FIG. 4 shows the long-lasting effects of Compound (1AA) and KPT-185. Real-time RT-PCR analysis of Hbb-bh1, Hbb-y, and Hbb-b1 mRNA levels in MEL cells 48 hours after removal of Compound (1AA) or KPT-185 from culture. The cells were first treated with Compound (1AA) or KPT-185 for 24 hours. Relative expression levels were calculated by normalizing to Rpl4 mRNA levels in the same sample and also to cells treated with DMSO.
  • FIG. 5 shows that KPT-185 is more effective than hydroxyurea in increasing embryonic hemoglobin expression in MEL cells. Real-time RT-PCR analysis of Hbb-bh1, Hbb-y, and Hbb-b1 mRNA levels in MEL cells after 48 hours of treatment with hydroxyurea (HU), KPT-185 (KPT), or control DMSO. Relative expression levels were calculated by normalizing to Rpl4 mRNA levels in the same sample and also to cells treated with DMSO.
  • FIG. 6 shows that Compound (1BB) increased embryonic and fetal hemoglobin expression levels in MEL cells. Real-time RT-PCR analysis of Hbb-bh1, Hbb-y and Hbb-b1 mRNA levels in MEL cells after 48 hours of treatment with compound (1BB) at indicated concentration or with vehicle water. Relative expression levels were calculated by normalizing to Rpl4 mRNA levels in the same sample and also to cells treated with water.
  • FIG. 7 shows KPT-330 and KPT-8602 increased embryonic and fetal hemoglobin expression levels in MEL cells. Real-time RT-PCR analysis of Hbb-bh1, Hbb-y and Hbb-b1 mRNA levels in MEL cells after 48 hours of treatment with KPT-330 and KPT-8602 at indicated concentration or with vehicle DMSO. Relative expression levels were calculated by normalizing to Rpl4 mRNA levels in the same sample and also to cells treated with DMSO.
  • FIG. 8 shows that Compound 1AA and 1BB significantly increased embryonic and fetal hemoglobin expression levels in human erythroid cells. Human CD34+ cells mobilized by granulocyte-colony stimulating factor (G-CSF) were induced to undergo erythroid differentiation in culture. Treatment with Compound 1AA and 1BB at indicated concentrations or vehicles (DMSO or water) were started at day 5 of expansion condition. Erythroid cells were harvested at day 10 of differentiation condition for real-time RT-PCR analysis of human embryonic s (FIG. 8A), fetal γ (FIG. 8B), and adult β (FIG. 8C) hemoglobin mRNA levels. Relative expression levels were calculated by normalizing to RPL4 mRNA levels in the same sample and also to cells treated with DMSO. (FIG. 8D) Representative bar graphs showing mRNA levels of human embryonic and fetal hemoglobin (γ+ε) as a percentage of total β-like globin (β+γ+ε) mRNA levels after treatment with Compound 1AA and 1BB versus DMSO. Note that a level of 20% or above for embryonic and fetal hemoglobin is considered curative for sickle cell disease. **, P<0.01; ***, P<0.001; ****P<0.0001 (two-tailed Student's t test).
  • DETAILED DESCRIPTION Definitions
  • The term “alkyl,” as used herein, means any straight chain or branched, non-cyclic or cyclic, unsaturated or saturated aliphatic hydrocarbon containing, for example, from 1 to 20 carbon atoms, while the term “lower alkyl” has the same meaning as alkyl but contains from 1 to 5 carbon atoms. The term “higher alkyl” has the same meaning as alkyl but contains from 6 to 20 carbon atoms. Representative saturated straight chain alkyls include, but are not limited to, methyl, ethyl, n-propyl, n-butyl, n-pentyl, n-hexyl, n-heptyl, n-octyl, n-nonyl, and the like; while saturated branched alkyls include, but are not limited to, isopropyl, sec-butyl, isobutyl, tert-butyl, isopentyl, and the like. Cyclic alkyls may be obtained by joining two alkyl groups bound to the same atom or by joining two alkyl groups each bound to adjoining atoms. Representative saturated cyclic alkyls include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like; while unsaturated cyclic alkyls include, but are not limited to, cyclopentenyl and cyclohexenyl, and the like. Cyclic alkyls are also referred to herein as “cycloalkyls,” “homocycles” or “homocyclic rings.” Unsaturated alkyls contain at least one double or triple bond between adjacent carbon atoms (referred to as an “alkenyl” or “alkynyl,” respectively). Representative straight chain and branched alkenyls include, but are not limited to, ethylenyl, propylenyl, 1-butenyl, 2-butenyl, isobutylenyl, 1-pentenyl, 2-pentenyl, 3-methyl-1-butenyl, 2-methyl-2-butenyl, 2,3-dimethyl-2-butenyl, and the like; while representative straight chain and branched alkynyls include, but are not limited to, acetylenyl, propynyl, 1-butynyl, 2-butynyl, 1-pentynyl, 2-pentynyl, 3-methyl-1-butynyl, and the like.
  • The term “aryl,” as used herein, refers to any aromatic carbocyclic moiety containing, for example, 5 to 20 carbon atoms such as, but not limited to, phenyl or naphthyl.
  • The term “arylalkyl,” or “aralkyl” as used herein, refers to any alkyl having at least one alkyl hydrogen atom replaced with an aryl moiety, such as benzyl, but not limited to, (CH2)2phenyl, —(CH2)3phenyl, —CH(phenyl)2, and the like.
  • The term “halogen” or “halo” as used herein, refers to any fluoro, chloro, bromo, or iodo moiety.
  • The term “haloalkyl” as used herein, refers to any alkyl having at least one hydrogen atom replaced with halogen, such as trifluoromethyl, and the like.
  • The term “heteroaryl” as used herein, refers to any aromatic heterocycle ring of 5 to 20 members and having at least one heteroatom selected from nitrogen, oxygen and sulfur, and containing at least one carbon atom, including, but not limited to, both mono- and bicyclic ring systems. Representative heteroaryls include, but are not limited to, furyl, benzofuranyl, thiophenyl, benzothiophenyl, pyrrolyl, indolyl, isoindolyl, azaindolyl, pyridyl, quinolinyl, isoquinolinyl, oxazolyl, isooxazolyl, benzoxazolyl, pyrazolyl, imidazolyl, benzimidazolyl, thiazolyl, benzothiazolyl, isothiazolyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, cinnolinyl, phthalazinyl, or quinazolinyl.
  • The term “heteroarylalkyl” as used herein, refers to any alkyl having at least one alkyl hydrogen atom replaced with a heteroaryl moiety, such as —CHpyridinyl, —CH2pyrimidinyl, and the like.
  • The term “heterocycle” or “heterocyclic,” “heterocyclyl” or “heterocyclic ring,” as used herein, refers to any non-aromatic 3- to 7-membered monocyclic or any non-aromatic 7- to 10-membered bicyclic heterocyclic ring which is either saturated or unsaturated and which contains from 1 to 4 heteroatoms independently selected from nitrogen, oxygen and sulfur, and wherein the nitrogen and sulfur heteroatoms may be optionally oxidized, and the nitrogen heteroatom may be optionally quaternized, including bicyclic rings in which any of the above heterocycles are fused to a benzene ring. The heterocycle may be attached via any heteroatom or carbon atom. Heterocycles may include, but are not limited to, morpholinyl, pyrrolidinonyl, pyrrolidinyl, piperidinyl, piperazinyl, imidazolinyl, imidazolidinyl, pyrazolinyl, pyrazolidinyl, 1,4-dioxanyl, dithianyl, hydantoinyl, valerolactamyl, oxiranyl, oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydropyridinyl, tetrahydroprimidinyl, tetrahydrothiophenyl, tetrahydrothiopyranyl, tetrahydropyrimidinyl, tetrahydrothiophenyl, tetrahydrothiopyranyl, and the like.
  • The term “heterocycloalkyl,” as used herein, refers to any alkyl having at least one alkyl hydrogen atom replaced with a heterocycle, such as —CH2morpholinyl, and the like.
  • The term “homocycle” or “cycloalkyl,” as used herein, refers to any saturated or unsaturated (non-aromatic) carbocyclic ring containing from 3-7 carbon atoms, such as, but not limited to, cyclopropane, cyclobutane, cyclopentane, cyclohexane, cycloheptane, cyclohexene, and the like.
  • The term “alkylamino,” as used herein, refers to at least one alkyl moiety attached through a nitrogen bridge (i.e., —N-(alkyl)N, such as a dialkylamino) including, but not limited to, methylamino, ethylamino, dimethylamino, diethylamino, and the like.
  • The term “alkyloxy” or “alkoxy”, as used herein, refers to any alkyl moiety attached through an oxygen bridge (i.e., —O-alkyl) such as, but not limited to, methoxy, ethoxy, and the like.
  • The term “alkylthio.” as used herein, refers to any alkyl moiety attached through a sulfur bridge (i.e., —S— alkyl) such as, but not limited to, methylthio, ethylthio, and the like.
  • The term “salts” as used herein, refers to any salt that complexes with identified compounds described herein. Examples of such salts include, but are not limited to, acid addition salts formed with inorganic acids (e.g., hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid, and the like), and salts formed with organic acids such as, but not limited to, acetic acid, oxalic acid, tartaric acid, succinic acid, malic acid, fumaric acid, maleic acid, ascorbic acid, benzoic acid, tannic acid, pamoic acid, alginic acid, polyglutamic, acid, naphthalene sulfonic acid, naphthalene disulfonic acid, and polygalacturonic acid. Salt compounds can also be administered as pharmaceutically acceptable quaternary salts known to a person skilled in the art, which specifically includes the quaternary ammonium salts of the formula —NRR′R″+Z—, wherein R, R′, R″ is independently hydrogen, alkyl, or benzyl, and Z is a counter ion, including, but not limited to, chloride, bromide, iodide, alkoxide, toluenesulfonate, methylsulfonate, sulfonate, phosphate, or carboxylate (such as benzoate, succinate, acetate, glycolate, maleate, malate, fumarate, citrate, tartrate, ascorbate, cinnamoate, mandeloate, and diphenylacetate). Salt compounds can also be administered as pharmaceutically acceptable pyridine cation salts having a substituted or unsubstituted partial formula: wherein Z is a counter ion, including, but not limited to, chloride, bromide, iodide, alkoxide, toluenesulfonate, methylsulfonate, sulfonate, phosphate, or carboxylate (such as benzoate, succinate, acetate, glycolate, maleate, malate, fumarate, citrate, tartrate, ascorbate, cinnamoate, mandeloate, and diphenylacetate).
  • The term, “pharmaceutically acceptable carrier” as used herein, includes any and all solvents, or a dispersion medium including, but not limited to, water, ethanol, a polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils, coatings, isotonic and absorption delaying agents, liposomes, commercially available cleansers, and the like. Supplementary bioactive ingredients also can be incorporated into such carriers.
  • The terms “reduce,” “inhibit,” “diminish,” “suppress,” “decrease,” “prevent” and grammatical equivalents (including “lower,” “smaller,” etc.) when in reference to the expression of any symptom in an untreated subject relative to a treated subject, mean that the quantity and/or magnitude of the symptoms in the treated subject is lower than in the untreated subject by any amount that is recognized as clinically relevant by any medically trained personnel. In various exemplary embodiments, the quantity and/or magnitude of the symptoms in the treated subject is at least 10% lower than, at least 25% lower than, at least 50% lower than, at least 75% lower than, and/or at least 90% lower than the quantity and/or magnitude of the symptoms in the untreated subject.
  • The term “induce”, “inducing”, “induction” and grammatical equivalents when in reference to the expression of genes or proteins in a treated subject or a treated cell relative to a untreated subject or a untreated cell, mean that the quantity and/or levels of the genes or proteins in the treated subject or treated cell is greater than that in the untreated subject or untreated cell by any amount that is recognized as clinically relevant by any medically trained personnel. In various exemplary embodiments, the quantity and/or levels of the genes or proteins in the treated subject or treated cell is at least 10% greater than, at least 25% greater than, at least 50% greater than, at least 75% greater than, and/or at least 90% greater than the quantity and/or levels of the genes or proteins in the untreated subject or untreated cell.
  • The term “inhibitory compound” as used herein, refers to any compound capable of interacting with (i.e., for example, attaching, binding etc.) to a binding partner under conditions such that the binding partner becomes unresponsive to its natural ligands. Inhibitory compounds may include, but are not limited to, small organic molecules, antibodies, and proteins/peptides.
  • The term “attached” as used herein, refers to any interaction between a medium (or carrier) and a drug. Attachment may be reversible or irreversible. Such attachment includes, but is not limited to, covalent bonding, ionic bonding, Van der Waals forces or friction, and the like. A drug is attached to a medium (or carrier) if it is impregnated, incorporated, coated, in suspension with, in solution with, mixed with, etc.
  • The term “drug” or “compound” as used herein, refers to any pharmacologically active substance capable of being administered which achieves a desired effect. Drugs or compounds can be synthetic or naturally occurring, non-peptide, proteins or peptides, oligonucleotides or nucleotides, polysaccharides or sugars.
  • The term “administered” or “administering,” as used herein, refers to any method of providing a composition to a patient such that the composition has its intended effect on the patient. An exemplary method of administering is by a direct mechanism such as, local tissue administration (i.e., for example, extravascular placement), oral ingestion, transdermal patch, topical, inhalation, suppository, etc.
  • The term “patient,” as used herein, is an animal, such as, for example, a mammal, such as, for example, a human that need not be hospitalized. For example, out-patients and persons in nursing homes are “patients.” A patient may comprise any age of a human or non-human animal and therefore includes both adult and juveniles (i.e., children). It is not intended that the term “patient” connote a need for medical treatment, therefore, a patient may voluntarily or involuntarily be part of experimentation whether clinical or in support of basic science studies.
  • The term “subject” as used herein refers to a vertebrate, preferably a mammal, more preferably a primate, still more preferably a human. Mammals include, without limitation, humans, primates, wild animals, feral animals, farm animals, sports animals and pets. The subject may be a juvenile or an adult.
  • Exportin 1” (“XPO1”), also known as chromosomal maintenance 1 (CRM1), is a protein that mediates the nuclear export of proteins and nucleic acids. In human, for instance, a representative sequence of the XPO1 protein is UniProt: O14980 (or RefSeq: NP_003391). The term “XPO1 inhibitor” refers to an agent that inhibits or reduces the expression or biological activity of XPO1. Example XPO1 inhibitors, whether antibodies, inhibitory nucleic acids, or small molecules, are well-known in the art. Specific XPO1 inhibitors are identified in, e.g., Q. Sun et al., Signal Transduct Target Ther. 1:16010 (2016); U.S. Pat. No. 9,861,615; and K. Parikh et al., J. of Hematol. & Oncol 7:78 (2014).
  • SET binding protein 1” (“SETBP1”) is a protein that binds SET and is a DNA-binding protein known to regulate gene transcription. In human, for instance, a representative sequence of the SETBP1 protein is UniProt: Q9Y6X0 (or RefSeq: NP_001123582, NP_056374, NP_001366070, or NP_001366071). The term “SETBP1 inhibitor” used herein refers to molecules that inhibit the activity of SETBP1 and/or inhibit the binding of SETBP1 to other proteins in a cell. Specific SETBP1 inhibitors, for example, are identified in WO2019/226773.
  • The term “therapeutically effective amount” or “effective amount” of a compound described herein or a pharmaceutically acceptable salt thereof means an amount sufficient to effect treatment when administered to a subject, to provide a therapeutic benefit such as amelioration of symptoms or slowing of disease progression. For example, a therapeutically effective amount may be an amount sufficient to decrease a symptom of a sickle cell disease. The therapeutically effective amount may vary depending on the subject, and disease or condition being treated, the weight and age of the subject, the severity of the disease or condition, and the manner of administering, which can readily be determined by one of ordinary skill in the art.
  • The instant inventors made the surprising and unexpected discovery, as the experimental examples demonstrated, that SETBP1 is a repressor of the transcription of the embryonic and fetal hemoglobin genes. In addition to the knock down experiments, small molecule SETBP1 inhibitors also demonstrated potent activation of embryonic and fetal hemoglobins.
  • In another surprising and unexpected finding, inhibition of XPO1 also led to significant activation of the embryonic and fetal hemoglobins, suggesting that XPO1 is a cofactor in the repression of embryonic and fetal hemoglobin transcription. These data therefore demonstrate that inhibition of SETBP1 and/or XPO1 can effectively treat diseases and conditions by increasing the production of embryonic and/or fetal hemoglobin.
  • An aspect of the invention is a method for treating a disease by activating the expression of embryonic hemoglobin in a subject, comprising administering to the subject a therapeutically effective amount of a SETBP1 inhibitor or an XPO1 inhibitor.
  • Another aspect of the invention is a method for treating a disease by activating expression of embryonic hemoglobin in a subject, comprising administering to a subject in need thereof a therapeutically effective amount of a SETBP1 inhibitor or a XPO1 inhibitor or a combination of a SETBP1 inhibitor and a XPO1 inhibitor.
  • Another aspect of the invention is a method for treating a disease by activating the expression of fetal hemoglobin in a subject, comprising administering to the subject a therapeutically effective amount of a SETBP1 inhibitor or an XPO1 inhibitor.
  • Another aspect of the invention is a method for treating a disease by activating expression of fetal hemoglobin in a subject, comprising administering to a subject in need thereof a therapeutically effective amount of a SETBP1 inhibitor or a XPO1 inhibitor or a combination of a SETBP1 inhibitor and a XPO1 inhibitor.
  • An exemplary embodiment of the invention is a method for treating sickle cell disease or β-thalassemia in a subject, comprising administering to the subject a therapeutically effective amount of a SETBP1 inhibitor.
  • Another exemplary embodiment of the invention is a method for treating sickle cell disease or D-thalassemia in a subject, comprising administering to the subject a therapeutically effective amount of an XPO1 inhibitor.
  • Another exemplary embodiment of the invention is a method for treating sickle cell disease or β-thalassemia in a subject, comprising administering to a subject in need thereof a therapeutically effective amount of a combination of a SETBP1 inhibitor and an XPO1 inhibitor.
  • Another exemplary embodiment of the invention is a method for treating sickle cell disease in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a SETBP1 inhibitor, an XPO1 inhibitor, or a combination of a SETBP1 inhibitor and an XPO1 inhibitor.
  • Another exemplary embodiment of the invention is a method for treating sickle cell disease (SCD) in a subject, comprising administering to a subject in need thereof a therapeutically effective amount of a SETBP1 inhibitor. Another exemplary embodiment of the invention is a method for treating sickle cell disease (SCD) in a subject, comprising administering to a subject in need thereof a therapeutically effective amount of a XPO1 inhibitor. Another exemplary embodiment of the invention is a method for treating sickle cell disease (SCD) in a subject, comprising administering to a subject in need thereof a therapeutically effective amount of a combination of a SETBP1 inhibitor and an XPO1 inhibitor.
  • In an exemplary embodiment, sickle cell disease is treated with a SETBP1 inhibitor.
  • In an exemplary embodiment, sickle cell disease is treated with an XPO1 inhibitor.
  • In an exemplary embodiment, sickle cell disease is treated with a combination of SETBP1 inhibitor and an XPO1 inhibitor.
  • In an exemplary embodiment, β-thalassemia is treated with a SETBP1 inhibitor.
  • In an exemplary embodiment, β-thalassemia is treated with an XPO1 inhibitor.
  • In an exemplary embodiment, β-thalassemia is treated with a combination of a SETBP1 inhibitor and an XPO1 inhibitor.
  • In an exemplary embodiment, the SETBP1 inhibitor is an shRNA or an siRNA. In an exemplary embodiment, the shRNA or the siRNA has a sequence that is at least 95% identical to the nucleic acid sequences selected from the group consisting of SEQ ID NO: 1 and SEQ ID NO: 2.
  • In an exemplary embodiment, the SETBP1 inhibitor is an anti-SETBP1 antibody inhibiting the activity of SETBP1 protein, a RNA molecule reducing SETBP1 transcription or translation, or a gene editing system disrupting the SETBP1 genomic sequence.
  • In an exemplary embodiment, the XPO1 inhibitor is an shRNA or an siRNA. In an exemplary embodiment, the XPO1 inhibitor is an anti-XPO1 antibody inhibiting the activity of XPO1 protein, a RNA molecule reducing SETBP1 transcription or translation, or a gene editing system disrupting the XPO1 genomic sequence.
  • In an exemplary embodiment, the SETBP1 inhibitor is a compound of Formula (1):
  • Figure US20220249486A1-20220811-C00001
  • or a pharmaceutically acceptable salt thereof, wherein
  • R1 is —OR8, —SR8 or —NR8R8;
  • R2, R3, R4, R5 and R6 are independently H or C1-4 alkyl;
  • R7 is —NR10R10 or a heterocyclyl group;
  • L is —(CR9R9)n—;
  • X is an organic or inorganic anion;
  • each R8 is independently H or C1-3 alkyl;
  • each R9 is independently H or C1-3 alkyl;
  • each R10 is independently H or C1-3 alkyl; and
  • n is 1, 2, 3 or 4.
  • In an exemplary embodiment of Formula (1), R1 is —OR8.
  • In an exemplary embodiment of Formula (1), R1 is —OR8 and R3 is H.
  • In an exemplary embodiment of Formula (1), R1 is —OR8, R3 is H, R8 is H and each R9 is H.
  • In an exemplary embodiment of Formula (1), n is 2.
  • In an exemplary embodiment of Formula (1), R1 is —OR8 and at least one R9 is H.
  • In an exemplary embodiment of Formula (1), R1 is —NR8R8, R3 is H, each R8 is H and each R9 is H.
  • In an exemplary embodiment of Formula (1), at least one of R4 and R5 is —CH3.
  • In an exemplary embodiment of Formula (1), each of R4 and R5 is —CH3.
  • In an exemplary embodiment of Formula (1), R1 is —OR8, R2 is H, R3 is H and each of R4 and R8 is —CH3.
  • In an exemplary embodiment of Formula (1), R7 is a heterocyclyl group, where the heterocyclyl group is a 5- or 6-membered ring containing 1 or 2 nitrogen atoms.
  • In an exemplary embodiment of Formula (1), R7 is a heterocyclyl group, R2 is H and R1 is —OR8.
  • In an exemplary embodiment of Formula (1), R7 is a heterocyclyl group, R2 is H, R1 is —OR8 and R3 is H.
  • In an exemplary embodiment of Formula (1), R7 is a heterocyclyl group where the hetereocyclyl group is a piperidinyl group, R1 is —OR8, R2 is H, R3 is H, R8 is H and each R9 is H.
  • In an exemplary embodiment of Formula (1), R7 is a heterocyclyl group where the hetereocyclyl group is a piperidinyl group, R2 is H and at least one R9 is H.
  • In an exemplary embodiment of Formula (1), R7 is a heterocyclyl group where the hetereocyclyl group is a piperidinyl group, R2 is H and each R9 is H.
  • In an exemplary embodiment of Formula (1), R7 is a heterocyclyl group where the hetereocyclyl group is a piperidinyl group, R2 is H and at least one of R4 and R8 is —CH3.
  • In an exemplary embodiment of Formula (1), R7 is a heterocyclyl group where the hetereocyclyl group is a piperidinyl group, R2 is H and each of R4 and R5 is —CH3.
  • In an exemplary embodiment of Formula (1), R7 is a heterocyclyl group where the hetereocyclyl group is a piperidinyl group, R1 is —OR8, R2 is H, R3 is H and each of R4 and R5 is —CH3.
  • In an exemplary embodiment of Formula (1), X is F, Cl, Br or I.
  • In an exemplary embodiment of Formula (1), R1 is a heterocyclyl group where the hetereocyclyl group is a piperidinyl group, R1 is —OR8, R2 is H, R3 is H, each of R4 and R5 is —CH3 and X is F, Cl, Br or I.
  • In an exemplary embodiment of Formula (1), R7 is —NR10R10.
  • In an exemplary embodiment of Formula (1), R7 is —NR10R10 and each R10 is independently C1-3 alkyl.
  • In an exemplary embodiment of Formula (1), R7 is —NR10R10 and one R10 is C1-3 alkyl and one R10 is —H.
  • In an exemplary embodiment of Formula (1), R2 is H, R7 is —NR10R10 and each R10 is —CH2CH3.
  • In an exemplary embodiment of Formula (1), R2 is H, R7 is —NR10R10 and each R10 is H.
  • In an exemplary embodiment of Formula (1), R2 is H, R7 is —NR10R10, R1 is —OR8 and R3 is H.
  • In an exemplary embodiment of Formula (1), R2 is H, R7 is —NR10R10, R1 is —OR8, R3 is H, R8 is H and each R9 is H.
  • In an exemplary embodiment of Formula (1), R2 is H, R7 is —NR10R10 and at least one R9 is H.
  • In an exemplary embodiment of Formula (1), R2 is H, R7 is —NR10R10 and each R9 is H.
  • In an exemplary embodiment of Formula (1), R7 is —NR10R10, each R10 is —CH2CH3 and at least one of R4 and R5 is —CH3.
  • In an exemplary embodiment of Formula (1), R2 is H, R7 is —NR10R10, each R10 is —CH2CH3 and each of R4 and R5 is —CH3.
  • In an exemplary embodiment of Formula (1), R2 is H, R7 is —NR10R10, each R10 is —CH2CH3, R1 is —OR8, R3 is H, each of R4 and R5 is —CH3 and X is F, Cl, Br or I.
  • In an exemplary embodiment of Formula (1), R7 is a heterocyclyl group selected from the group consisting of morpholinyl, pyrrolidinonyl, pyrrolidinyl, piperidinyl, piperazinyl and tetrahydropyranyl.
  • In an exemplary embodiment of Formula (1), R7 is a heterocyclyl group selected from the group consisting of morpholinyl, piperidinyl, piperazinyl and tetrahydropyranyl.
  • In an exemplary embodiment of the compound of Formula (1), R1 is —OR8; R2, R3 and R6 are H; R4 and R5 are independently C1-3 alkyl; R7 is —NR10R10 or a piperidinyl group; L is —(CR9R9)n—; X is an organic or inorganic anion; R8 is H or C1-3 alkyl; each R9 is independently H or C1-3 alkyl; each R10 is independently H or C1-3 alkyl; and n is 1, 2, 3 or 4.
  • In a particular embodiment, X is selected from the group consisting of F, Cl, Br and I. In another particular embodiment, n is 2.
  • In an exemplary embodiment of the compound of Formula (1), R1 is —OR8; R2, R3, R6, R8 and R9 are H; R4 and R5 are —CH3; R7 is a piperidinyl group; L is —(CR9R9)n—; X is selected from the group consisting of F, Cl, Br and I; and n is 1, 2, 3 or 4.
  • In an exemplary embodiment of the compound of Formula (1), R1 is —OR8; R2, R3, R6, R8 and R9 are H; R4 and R5 are —CH3; R7 is —NR10R10; each R10 is independently C1-3 alkyl; L is —(CR9R9)n—; X is selected from the group consisting of F, Cl, Br and I; and n is 1, 2, 3 or 4.
  • In an exemplary embodiment, the compound of Formula (1) is a compound of Formula (1A):
  • Figure US20220249486A1-20220811-C00002
  • wherein X is an organic or inorganic anion. In an exemplary embodiment, X is selected from the group consisting of halo (F, Cl, Br, I), alkoxide, p-toluenesulfonate, methylsulfonate, sulfonate, phosphate, and carboxylate (such as benzoate, succinate, acetate, glycolate, maleate, malate, fumarate, citrate, tartrate, ascorbate, cinnamoate, mandeloate, and diphenylacetate). In a particular embodiment, X is selected from the group consisting of F, Cl, Br, and I.
  • In an exemplary embodiment, the compound of Formula (1) is Compound (1AA):
  • Figure US20220249486A1-20220811-C00003
  • In an exemplary embodiment, the compound of Formula (1) is a compound of Formula (1B):
  • Figure US20220249486A1-20220811-C00004
  • wherein X is an organic or inorganic anion. In an exemplary embodiment, X is selected from the group consisting of halo (F, Cl, Br, I), alkoxide, p-toluenesulfonate, methylsulfonate, sulfonate, phosphate, and carboxylate (such as benzoate, succinate, acetate, glycolate, maleate, malate, fumarate, citrate, tartrate, ascorbate, cinnamoate, mandeloate, and diphenylacetate). In a particular embodiment, X is selected from the group consisting of F, Cl, Br and I.
  • In an exemplary embodiment, the compound of Formula (1) is Compound (1BB):
  • Figure US20220249486A1-20220811-C00005
  • In an exemplary embodiment, the SETBP1 inhibitor is a compound of Formula (2):
  • Figure US20220249486A1-20220811-C00006
  • or a pharmaceutically acceptable salt thereof, wherein
  • R1 is an aryl group or a heteroaryl group, where the aryl group and the heteroaryl group each has 0 to 3 substituents independently selected from the group consisting of —NR6R6, OH, F, Cl, Br and C1-3 alkyl;
  • R2 and R3 are independently H or C1-6 alkyl;
  • R4 and R5 are independently H, —NR6R6 or OR6;
  • each R6 is independently H or C1-3 alkyl;
  • X1 and X2 are independently CH or N;
  • L is —(CR7R7)n—;
  • each R7 is independently H or C1-3 alkyl; and
  • n is 1, 2, 3 or 4.
  • In an exemplary embodiment of Formula (2), the aryl or heteroaryl group of R1 is selected from the group consisting of phenyl, furyl, benzofuranyl, thiophenyl, benzothiophenyl, pyrrolyl, pyridyl and pyrimidinyl.
  • In an exemplary embodiment of Formula (2), R1 is a phenyl group having 1 substituent that is —NR6R6.
  • In an exemplary embodiment of Formula (2), R1 is a phenyl group having 1 substituent that is —NR6R6 where at least one of R6 is H.
  • In an exemplary embodiment of Formula (2), R1 is a phenyl group having 1 substituent that is —NR6R6 where each R6 is H.
  • In an exemplary embodiment of Formula (2), at least one of R2 and R3 is H.
  • In an exemplary embodiment of Formula (2), each of R2 and R3 is H.
  • In an exemplary embodiment of Formula (2), X1 and X2 are each N and at least one of R4 and R5 is —NR6R6.
  • In an exemplary embodiment of Formula (2), X1 and X2 are each N and each of R4 and R5 is —NR6R6.
  • In an exemplary embodiment of Formula (2), X1 and X2 are each N and at least one of R4 and R5 is —NR6R6 where each R6 is H.
  • In an exemplary embodiment of Formula (2), X1 and X2 are each N and each of R4 and R5 is —NR6R6 where each R6 is H.
  • In an exemplary embodiment of Formula (2), L is —(CR7R7)— and each R7 is independently H or C1-3 alkyl. In an exemplary embodiment of Formula (2), L is —CH2—.
  • In an exemplary embodiment, the compound of Formula (2) is a compound of Formula (2A):
  • Figure US20220249486A1-20220811-C00007
  • wherein the compound of Formula (2A) exists as a pharmaceutically acceptable salt, wherein HA is a proton donor. In an exemplary embodiment, HA is selected from the group consisting of inorganic acids (e.g., hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid, and the like) and organic acids (e.g., acetic acid, oxalic acid, tartaric acid, succinic acid, malic acid, fumaric acid, maleic acid, ascorbic acid, benzoic acid, tannic acid, pamoic acid, alginic acid, polyglutamic, acid, naphthalene sulfonic acid, naphthalene disulfonic acid and polygalacturonic acid). In a particular embodiment, HA is selected from the group consisting of hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid and nitric acid.
  • In an exemplary embodiment, the compound of Formula (2) is Compound (2AA):
  • Figure US20220249486A1-20220811-C00008
  • Compounds as described herein may be synthesized according to methods known in the art or may be commercially available.
  • In an exemplary embodiment, the compound of Formula (1), Formula (1A), Compound (1AA), Formula (1B), or Compound (1BB) is present in a pharmaceutical composition comprising one or more excipients.
  • In an exemplary embodiment, the compound of Formula (1) is present in a pharmaceutical composition comprising one or more excipients.
  • In an exemplary embodiment, the compound of Formula (1A) is present in a pharmaceutical composition comprising one or more excipients.
  • In an exemplary embodiment. Compound (1AA) is present in a pharmaceutical composition comprising one or more excipients.
  • In an exemplary embodiment, the compound of Formula (1B) is present in a pharmaceutical composition comprising one or more excipients.
  • In an exemplary embodiment. Compound (1BB) is present in a pharmaceutical composition comprising one or more excipients.
  • In an exemplary embodiment, the compound of Formula (2), Formula (2A), or Compound (2AA) is present in a pharmaceutical composition comprising one or more excipients.
  • In an exemplary embodiment, the compound of Formula (2) is present in a pharmaceutical composition comprising one or more excipients.
  • In an exemplary embodiment, the compound of Formula (2A) is present in a pharmaceutical composition comprising one or more excipients.
  • In an exemplary embodiment. Compound (2AA) is present in a pharmaceutical composition comprising one or more excipients.
  • In an exemplary embodiment, a SETBP1 inhibitor and an XPO1 inhibitor are present in a pharmaceutical composition comprising one or more excipients. In an exemplary embodiment, the SETBP1 inhibitor or the XPO1 inhibitor or the combination of the SETBP1 inhibitor and the XPO1 inhibitor is present in a pharmaceutical composition comprising one or more excipients.
  • In an exemplary embodiment, the XPO1 inhibitor is selected from the group consisting of KPT-185, KPT-276, KPT-330, KPT-335, KPT-8602, and combination thereof. In an exemplary embodiment, the XPO1 inhibitor is selected from the group consisting of KPT-185, KPT-276, KPT-330, KPT-335 and KPT-8602.
  • An exemplary embodiment of the invention is a method for treating sickle cell disease (SCD) or β-thalassemia in a subject, comprising administering to the subject a therapeutically effective amount of a compound of Formula (1) (such as a compound of Formula (1A), such as Compound (1AA), such as a compound of Formula (1), such as Compound (1 TB)) or a compound of Formula (2) (such as a compound of Formula (2A), such as Compound (2AA)).
  • An exemplary embodiment of the invention is a method for treating sickle cell disease (SCD) or β-thalassemia in a subject, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of Formula (1) (such as a compound of Formula (1A), such as Compound (1AA), or such as a compound of Formula (1B), such as Compound (1B)).
  • An exemplary embodiment of the invention is a method for treating sickle cell disease (SCD) or β-thalassemia in a subject, comprising administering to a subject in need thereof a therapeutically effective amount of a combination of a compound of Formula (1) (such as a compound of Formula (1A), such as Compound (1AA), or such as a compound of Formula (1B), such as Compound (1B)) and a XPO1 inhibitor.
  • An exemplary embodiment of the invention is a method for treating sickle cell disease (SCD) or β-thalassemia in a subject, comprising administering to a subject in need thereof a therapeutically effective amount of a combination of a compound of Formula (1) (such as a compound of Formula (1A), such as Compound (1AA), or such as a compound of Formula (1B), such as Compound (1B)) and a XPO1 inhibitor selected from the group consisting of KPT-185, KPT-276, KPT-330, KPT-335, KPT-8602 and combinations thereof.
  • An exemplary embodiment of the invention is a method for treating sickle cell disease or β-thalassemia in a subject, comprising administering to the subject a therapeutically effective amount of a combination of a compound of Formula (1) (such as a compound of Formula (1A), such as Compound (1AA), or such as a compound of Formula (1B), such as Compound (1BB)) or a compound of Formula (2) (such as a compound of Formula (2A), such as Compound (2AA)) and an XPO1 inhibitor selected from the group consisting of KPT-185, KPT-276, KPT-330, KPT-335, KPT-8602 and combinations thereof.
  • Another exemplary embodiment of the invention is a method for treating sickle cell disease or β-thalassemia in a subject, comprising administering to the subject a therapeutically effective amount of a XPO1 inhibitor selected from the group consisting of KPT-185, KPT-276, KPT-330, KPT-335, KPT-8602 and combinations thereof.
  • Another exemplary embodiment of the invention is a method for treating sickle cell disease (SCD) in a subject, comprising administering to a subject in need thereof a therapeutically effective amount of a XPO1 inhibitor selected from the group consisting of KPT-185, KPT-276, KPT-330, KPT-335, KPT-8602 and combinations thereof.
  • Another exemplary embodiment of the invention is a method for treating β-thalassemia in a subject, comprising administering to the subject a therapeutically effective amount of a SETBP1 inhibitor or a combination of a SETBP1 inhibitor and an XPO1 inhibitor. Another exemplary embodiment of the invention is a method for treating 3-thalassemia in a subject, comprising administering to the subject a therapeutically effective amount of a SETBP1 inhibitor. Another exemplary embodiment of the invention is a method for treating β-thalassemia in a subject, comprising administering to the subject a therapeutically effective amount of a combination of a SETBP1 inhibitor and an XPO1 inhibitor.
  • In some embodiments described herein, wherein the subject is an adult human subject or a juvenile human subject. In an exemplary embodiment, the human subject is an adult human subject. In an exemplary embodiment, the human subject is a juvenile human subject.
  • Some embodiments provides for a method of inducing embryonic and fetal hemoglobin expression in a cell, wherein the cell is a hematopoietic progenitor cell, an erythroid progenitor cell, or an erythroid cell, comprising contacting the cell with a SETBP1 inhibitor, an XPO1 inhibitor, or a combination of a SETBP1 inhibitor and an XPO1 inhibitor.
  • In an exemplary embodiment, the invention provides a pharmaceutical composition comprising at least one pharmaceutically acceptable carrier, in addition to one or more of the compounds described herein. The composition can be present in any suitable form for the desired route of administration. Where the composition is to be administered orally, any suitable orally deliverable dosage form can be used, including, without limitation, tablets, capsules (solid or liquid filled), powders, granules, syrups and other liquids, elixirs, inhalants, troches, lozenges and solutions. Injectable compositions or i.v. infusions are also provided in the form of solutions, suspensions, and emulsions.
  • The compounds of the invention can be formulated as described herein and are suitable for administration in a therapeutically effective amount to the subject in any number of ways. A therapeutically effective amount of a compound as described herein depends upon the amounts and types of excipients employed, the amounts and specific types of active ingredients present in a dosage form, and the route by which the compound is to be administered to patients.
  • Typical dosage levels for the compounds of the invention generally range from about 0.001 to about 100 mg per kg of the subject's body weight per day which can be administered in single or multiple doses. An exemplary dosage is about 0.01 to about 25 mg/kg per day or about 0.05 to about 10 mg/kg per day. In other exemplary embodiments, the dosage level ranges from about 0.01 to about 25 mg/kg per day, such as about 0.05 to about 10 mg/kg per day, or about 0.1 to about 5 mg/kg per day.
  • A dose can typically range from about 0.1 mg to about 2000 mg per day and can be given as a single once-a-day dose or, alternatively, as divided doses throughout the day, optionally taken with food. In a particular embodiment, the daily dose is administered twice daily in equally divided doses. A daily dose range can range from about 5 mg to about 500 mg per day such as, for example, between about 10 mg and about 300 mg per day. In managing the patient, the therapy can be initiated at a lower dose, such as from about 1 mg to about 25 mg, and increased if necessary up to from about 200 mg to about 2000 mg per day as either a single dose or divided doses, depending on the subject's global response.
  • Suitable oral compositions in accordance with the invention include, without limitation, tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, syrups or elixirs. Inventive compositions suitable for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions. For example, liquid formulations of the compounds can contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations of the active agents.
  • For tablet compositions, typical non-toxic pharmaceutically acceptable excipients include, without limitation, inert diluents such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents such as, for example, corn starch, or alginic acid; binding agents such as, for example, starch, gelatin or lubricating agents such as, for example, magnesium stearate, stearic acid or talc. The tablets may be uncoated or, alternatively, they may be coated by known coating techniques to delay disintegration and absorption in the gastrointestinal tract and thereby to provide a sustained therapeutic action over a desired time period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate may be employed.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent such as, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium such as, for example peanut oil, liquid paraffin or olive oil.
  • For aqueous suspensions, the compound is admixed with excipients suitable for maintaining a stable suspension. Examples of such excipients include, without limitation, sodium carboxymethylcellulose, methylcellulose, hydropropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia.
  • Oral suspensions can also contain dispersing or wetting agents, such as naturally-occurring phosphatide such as, for example, lecithin, or condensation products of an alkylene oxide with fatty acids such as, for example, polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols such as, for example, heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as, for example, polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides such as, for example, polyethylene sorbitan monooleate. The aqueous suspensions may also contain one or more preservatives such as, for example, ethyl or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents such as sucrose or saccharin.
  • Sweetening agents such as those set forth above, and flavoring agents may be added to provide palatable oral preparations. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water can provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients such as, for example, sweetening, flavoring and coloring agents, may also be present.
  • Syrups and elixirs may be formulated with sweetening agents such as, for example, glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, and flavoring and coloring agents.
  • Compositions for parenteral administrations are formulated in a sterile medium suitable for intravenous, intramuscular or intrathecal delivery. A sterile injectable preparation of the compounds may be in the form of a sterile injectable solution or sterile injectable suspension. Non-toxic, parentally acceptable diluents or solvents such as, for example, 1,3-butanediol can be used to formulate the parenteral compositions. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile oils also can be employed as a solvent or a suspending medium. For this purpose, any bland fixed oil may be employed, including synthetic monoglycerides or diglycerides. In addition, fatty acids such as oleic acid can be used in the preparation of injectables.
  • Depending on the vehicle used and the concentration of the drug in the formulation, the parenteral formulation can contain other adjuvants such as local anesthetics, preservatives and buffering agents.
  • In addition to using shRNA technology, siRNA technology, antibodies, or small molecule inhibitors of SETBP1 or XPO1, it is reasonable for a person of skilled in the art to use gene editing technologies to inhibit the expression levels of SETBP1 or XPO1 and as a result to enhance the expression levels of embryonic and fetal hemoglobins. The editing, for instance, can introduce a mutation to the active site, a premature stop codon or a frameshift mutation into the SETBP1 or XPO1 gene. Gene editing technologies may include, but are not limited to, a CRISPR/Cas9 system, a CRISPR/Cas13 system, a zinc finger nuclease system, a TALEN system, and a meganuclease system. These gene editing technologies themselves are described in public domain (e.g. U.S. Pat. No. 10,196,652, Science: Vol. 358, Issue 6366, pp. 1019-1027. DOI: 10.1126/science.aaq0180).
  • In some embodiments, the SETBP1 inhibitor is one or more components of a gene editing system targeting one or more sites within a gene encoding SETBP1 or a regulatory element thereof, a nucleic acid molecule encoding the one or more components of the gene editing system, or a combination thereof. In some embodiments, the SETBP1 inhibitor is a gene editing system, and wherein the gene editing system is selected from the group consisting of CRISPR/Cas9, CRISPR/Cas13, a zinc finger nuclease system, a TALEN system, and a meganuclease system. In some embodiments, the gene editing system is a CRISPR/Cas9 system. In some embodiments, the gene editing system is a CRISPR/Cas13 system. In some embodiments, the gene editing system is a zinc finger nuclease system. In some embodiments, the gene editing system is a TALEN system. In some embodiments, the gene editing system is a meganuclease system. In some embodiments, the SETBP1 inhibitor comprises a guide RNA molecule comprising a tracr and a crRNA. In some embodiments, the crRNA comprises a targeting domain that is complementary with a target sequence of SETBP1.
  • In some embodiments, the XPO1 inhibitor is one or more components of a gene editing system targeting one or more sites within a gene encoding XPO1 or a regulatory element thereof, a nucleic acid molecule encoding the one or more components of the gene editing system, or a combination thereof. In some embodiments, the XPO1 inhibitor is a gene editing system, and wherein the gene editing system is selected from the group consisting of CRISPR/Cas9, CRISPR/Cas13, a zinc finger nuclease system, a TALEN system, and a meganuclease system. In some embodiments, the gene editing system is a CRISPR/Cas9 system. In some embodiments, the gene editing system is a CRISPR/Cas13 system. In some embodiments, the gene editing system is a zinc finger nuclease system. In some embodiments, the gene editing system is a TALEN system. In some embodiments, the gene editing system is a meganuclease system. In some embodiments, the XPO1 inhibitor comprises a guide RNA molecule comprising a tracr and a crRNA. In some embodiments, the crRNA comprises a targeting domain that is complementary with a target sequence of XPO1.
  • The pharmaceutical compositions according to the invention may contain one or more additional therapeutic agents, for example, to increase efficacy and/or to decrease side effects. Examples of such agents include, without limitation, agents to treat or inhibit immunological, inflammatory, autoimmune or allergic disorders.
  • EXAMPLES Protocol
  • For studies using MEL cells, the cells were treated with selected inhibitors or vehicle in a culture medium (RPM 11640 medium with 10% fetal bovine serum) for 48 hours and were subsequently harvested for analysis of relative mRNA levels of Hbb-bh1, Hbb-y, and Hbb-b1 using real-time RT-PCR. For studies using primary human CD34+ hematopoietic stem and progenitor cells, the cells were treated with inhibitors or vehicle following or concurrent with induction of erythroid differentiation using an established two-phase culture protocol (Xu et al., Genes & Development 24(8): 783-798 (2010)). Relative mRNA levels of β-like globin genes in human cells after treatment were also analyzed using real-time RT-PCR.
  • The non-targeting control pLKO.1 lentiviral shRNA construct shNC was purchased from Sigma (SHC002). Lentiviral shRNA constructs targeting Setbp1 (sh84 and sh8) were generated by cloning Setbp1 targeting sequences (sh84, CGG CTT TGA ATC CCA ATC ATT (SEQ ID NO:1); sh8, CCT ATG ATG AAC CTT GGT TAT (SEQ ID NO:2)) into pLKO.1. To generate infectious lentivirus, the constructs were co-transfected using FuGENE 6 transfection reagent into 293T cells along with packaging plasmid A8.9 and a plasmid expressing VSV-G, and virus were harvested at 72 hours after transfection. Viral titers were calculated by infecting NIH-3T3 cells with serial dilutions of viral stocks and counting puromycin resistant colonies. Lentiviral infections were performed by spinoculation in which a mixture of lentivirus and MEL cells at 4:1 ratio in 24-well plates were spun at 2000×g for 90 minutes at 37° C. Puromycin (2 μg/mL) was added to culture at 24 hours after spinoculation to select for infected cells.
  • Results
  • To evaluate whether Setbp1 plays a role in the repression of embryonic and fetal hemoglobin expression, the expression levels of Setbp1 were knocked down using two different lentiviral shRNAs in MEL cells, which is a mouse erythroid progenitor cell line conventionally used for studying repression of embryonic and fetal hemoglobins. Unexpectedly, the Setbp1 knockdowns significantly increased the mRNA levels of embryonic (Hbb-bh1) and fetal (Hbb-y) hemoglobin genes (FIG. 1), suggesting that Setbp1 is a repressor of their transcription.
  • Compound (1AA), which has been identified as an inhibitor of SETBP1 transcriptional activity (WO2019/226773), was tested for its effect on the expression of Hbb-bh1 and Hbb-v in MEL cells. Consistent with Setbp1 knockdown studies, treatment with Compound (1AA) at 0.1 μM for 48 hours led to over an 8-fold increase in the mRNA levels of both Hbb-bh1 and Hbb-y (FIG. 2). This unexpectedly potent activating effect of Compound (1AA) appears limited to embryonic and fetal hemoglobin since only minor changes in the levels of adult hemoglobin Hbb-b1 were detected under the same conditions (FIG. 2). Similarly, Compound (1BB), another inhibitor of SETBP1 (WO2019/226773), was also observed to significantly increase the expression of Hbb-bh1 and Hbb-y in MEL cells while having minor effects on Hbb-b1 mRNA levels (FIG. 6).
  • Compound (1AA) has been shown to inhibit the activity of SETBP1 by blocking its interaction with key cofactor XPO1 (WO2019/226773). This interaction between SETBP1 and XPO1 can also be blocked by XPO1 inhibitors such as, but not limited to, KPT-185 and KPT-330. Therefore, XPO1 inhibitors were also tested for their effect on the expression of Hbb-bh1 and Hbb-y in MEL cells. Treatment with 0.1 μm KPT-185 for 48 hours was observed to increase mRNA levels of Hbb-bh1 and Hbb-y by over 60-fold and over 14-fold, respectively (FIG. 3). Similarly, two other XPO1 inhibitors KPT-330 and KPT-8602 also were found to significantly increase mRNA levels of Hbb-bh1 and Hbb-y in MEL cells (FIG. 7). These results clearly show that XPO1 inhibitors are also surprisingly efficient in activating embryonic and fetal hemoglobin expression. Similar to Compounds (1AA) and (1BB), XPO1 inhibitors also a exerted minimal effect on the expression levels of Hbb-b1 (FIG. 2 and FIG. 7).
  • A further experiment examined whether continuous treatment of Compound (1AA) and KPT-185 as exemplary SETBP1 and XPO1 inhibitors, respectively, was required for activation of Hbb-bh1 and Hbb-y expression. MEL cells were treated with Compound (1AA) and KPT-185 for 24 hours and the Hbb-bh1 and Hbb-y mRNA levels were then examined at 48 hours after their removal. Significantly elevated levels of Hbb-bh1 and Hbb-y mRNA were still detected in MEL cells (FIG. 4). These results show a long-lasting activation effect of Compound (1AA) and KPT-185 on Hbb-bh1 and Hbb-y expression.
  • The ability of KPT-185 to activate Hbb-bh1 and Hbb-y expression in MEL cells was also compared to that of hydroxyurea, an FDA-approved drug for sickle cell disease. While KPT-185 was observed to activate Hbb-y expression to similar levels to that of hydroxyurea, surprisingly KPT-185 was substantially more effective than hydroxyurea in activating Hbb-bh1 expression with almost 30-fold higher levels of Hbb-bh1 mRNA being detected in cells treated with KPT-185 compared to cells treated with hydroxyurea (FIG. 5). This finding shows that XPO1 inhibitors such as KPT-185 are more effective than hydroxyurea in treating sickle cell disease.
  • Erythroid Differentiation and Treatment of Human CD34+ Cells
  • Primary human mobilized CD34+ cells were initially cultured in expansion medium containing StemSpan SFEM Medium (StemCell Technologies Inc.) with 1×CC100 cytokine mix (StemCell Technologies Inc.) and 2% Penicillin-Streptomycin (Life Technologies Inc.). Cells were maintained in this expansion medium at a density of 0.1-1×106 cells/mL for a total of six days with media changes every two days. On day 6, cells were transferred into differentiation medium containing StemSpan SFEM Medium with 2% Penicillin-Streptomycin, 20 ng/ml SCF (Biolegend), 1 U/ml EPO (Amgen), 5 ng/ml IL-3 (Biolegend), 2 μM dexamethasone (Sigma), and 1 μM β-estradiol (Sigma). Cells were maintained at a density of 0.1-1×106 cells/mL in differentiation medium for 10 days with media changes every two days. Drug treatments were started at day 5 of expansion condition and maintained thereafter. Fresh drugs were added at each medium change. The data is summarized in FIG. 8.
  • All patents and publications cited herein are incorporated by reference in their entireties.

Claims (58)

What is claimed is:
1. A method for treating sickle cell disease in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a SETBP1 inhibitor, an XPO1 inhibitor, or a combination of a SETBP1 inhibitor and an XPO1 inhibitor.
2. The method of claim 1, wherein the subject is an adult human subject or a juvenile human subject.
3. The method of claim 1, wherein the SETBP1 inhibitor is a compound of Formula (1):
Figure US20220249486A1-20220811-C00009
wherein:
R1 is —OR8;
R2, R3, and R6 are H;
R4 and R5 are independently C1-4 alkyl;
R7 is —NR10R10 or a piperidinyl group;
L is —(CR9R9)n—;
X is an organic or inorganic anion;
R8 is H or C1-3 alkyl;
each R9 is independently H or C1-3 alkyl;
each R10 is independently H or C1-3 alkyl; and
n is 1, 2, 3 or 4.
4. The method of claim 3, wherein X is selected from the group consisting of F, Cl, Br, and I.
5. The method of claim 3, wherein
R1 is —OR8;
R2, R3, R6, R8, and R9 are H;
R4 and R5 are —CH3;
R7 is a piperidinyl group;
L is —(CR9R9)n—;
X is selected from the group consisting of F, Cl, Br, and I; and
n is 1, 2, 3 or 4.
6. The method of claim 3, wherein
R1 is —OR8;
R2, R3, R6, R8 and R9 are H;
R4 and R5 are —CH3;
R7 is —NR10R10;
each R10 is independently C1-3 alkyl;
L is —(CR9R9)n—;
X is selected from the group consisting of F, Cl, Br, and I; and
n is 1, 2, 3 or 4.
7. The method of claim 3, where the compound of Formula (1) is a compound of Formula (1A)
Figure US20220249486A1-20220811-C00010
wherein X is selected from the group consisting of F, Cl, Br, and I.
8. The method of claim 3, where the compound of Formula (1) is Compound (1AA)
Figure US20220249486A1-20220811-C00011
9. The method of claim 3, wherein the compound of Formula (1) is a compound of Formula (1B)
Figure US20220249486A1-20220811-C00012
wherein X is selected from the group consisting of F, Cl, Br, and I.
10. The method of claim 3, wherein the compound of Formula (1) is Compound (1BB)
Figure US20220249486A1-20220811-C00013
11. The method of claim 1, wherein the SETBP1 inhibitor is a compound of Formula (2A)
Figure US20220249486A1-20220811-C00014
wherein HA is a proton donor.
12. The method of claim 11, wherein HA is selected from the group consisting of hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, and nitric acid.
13. The method of any one of claims 11-12, wherein the compound of Formula (2A) is Compound (2AA)
Figure US20220249486A1-20220811-C00015
14. The method of claim 1, wherein the SETBP1 inhibitor is an shRNA or an siRNA.
15. The method of claim 14, wherein the shRNA or the siRNA has a sequence that is at least 95% identical to a nucleic acid sequence selected from the group consisting of SEQ ID NO: 1 and SEQ ID NO: 2.
16. The method of claim 1, wherein the SETBP1 inhibitor is a gene editing system, and wherein the gene editing system is selected from the group consisting of CRISPR/Cas9, CRISPR/Cas13, a zinc finger nuclease system, a TALEN system, and a meganuclease system.
17. The method of claim 1, wherein the XPO1 inhibitor is selected from the group consisting of KPT-185, KPT-276, KPT-330, KPT-335 and KPT-8602.
18. The method of claim 1, wherein the XPO1 inhibitor is an shRNA or an siRNA.
19. The method of claim 1, wherein the XPO1 inhibitor is a gene editing system, and wherein the gene editing system is selected from the group consisting of CRISPR/Cas9, CRISPR/Cas13, a zinc finger nuclease system, a TALEN system, and a meganuclease system.
20. The method of claim 1, wherein the SETBP1 inhibitor or the XPO1 inhibitor or the combination of the SETBP1 inhibitor and the XPO1 inhibitor is present in a pharmaceutical composition comprising one or more excipients.
21. A method for treating β-thalassemia in a subject, comprising administering to the subject a therapeutically effective amount of a SETBP1 inhibitor, or a combination of a SETBP1 inhibitor and an XPO1 inhibitor.
22. The method of claim 21, wherein the subject is an adult human subject or a juvenile human subject.
23. The method of claim 21, wherein the SETBP1 inhibitor is a compound of Formula (1):
Figure US20220249486A1-20220811-C00016
wherein:
R1 is —OR8;
R2, R3, and R6 are H;
R4 and R5 are independently C1-4 alkyl;
R7 is —NR10R10 or a piperidinyl group;
L is —(CR9R9)n—;
X is an organic or inorganic anion;
R8 is H or C1-3 alkyl;
each R9 is independently H or C1-3 alkyl;
each R10 is independently H or C1-3 alkyl; and
n is 1, 2, 3 or 4.
24. The method of claim 23, wherein X is selected from the group consisting of F, Cl, Br, and I.
25. The method of claim 23, wherein
R1 is —OR8;
R2, R3, R6, R8, and R9 are H;
R4 and R5 are —CH3;
R7 is a piperidinyl group;
L is —(CR9R9)n—;
X is selected from the group consisting of F, Cl, Br, and I; and
n is 1, 2, 3 or 4.
26. The method of claim 23, wherein
R1 is —OR8;
R2, R3, R6, R8 and R9 are H;
R4 and R5 are —CH3;
R7 is —NR10R10;
each R10 is independently C1-3 alkyl;
L is —(CR9R9)n—;
X is selected from the group consisting of F, Cl, Br, and I; and
n is 1, 2, 3 or 4.
27. The method of claim 23, wherein the compound of Formula (1) is a compound of
Figure US20220249486A1-20220811-C00017
wherein X is selected from the group consisting of F, Cl, Br, and I.
28. The method of claim 23, wherein the compound of Formula (1) is Compound (1AA)
Figure US20220249486A1-20220811-C00018
29. The method of claim 23, wherein the compound of Formula (1) is a compound of Formula (1B)
Figure US20220249486A1-20220811-C00019
wherein X is selected from the group consisting of F, Cl, Br, and I.
30. The method of claim 23, wherein the compound of Formula (1) is Compound (1BB)
Figure US20220249486A1-20220811-C00020
31. The method of claim 21, wherein the SETBP1 inhibitor is a compound of Formula (2A)
Figure US20220249486A1-20220811-C00021
wherein HA is a proton donor.
32. The method of claim 31, wherein HA is selected from the group consisting of hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, and nitric acid.
33. The method of any one of claims 31-32, wherein the compound of Formula (2A) is Compound (2AA)
Figure US20220249486A1-20220811-C00022
34. The method of claim 21, wherein the SETBP1 inhibitor is an shRNA or an siRNA.
35. The method of claim 34, wherein the shRNA or the siRNA has a sequence that is at least 95% identical to a nucleic acid sequence selected from the group consisting of SEQ ID NO: 1 and SEQ ID NO: 2.
36. The method of claim 21, wherein the SETBP1 inhibitor is a gene editing system, and wherein the gene editing system is selected from the group consisting of CRISPR/Cas9, CRISPR/Cas13, a zinc finger nuclease system, a TALEN system, and a meganuclease system.
37. The method of claim 21, wherein the XPO1 inhibitor is selected from the group consisting of KPT-185, KPT-276, KPT-330, KPT-335 and KPT-8602.
38. The method of claim 21, wherein the XPO1 inhibitor is an shRNA or an siRNA.
39. The method of claim 21, wherein the XPO1 inhibitor is a gene editing system, and wherein the gene editing system is selected from the group consisting of CRISPR/Cas9, CRISPR/Cas13, a zinc finger nuclease system, a TALEN system, and a meganuclease system.
40. The method of claim 21, wherein the SETBP1 inhibitor or the XPO1 inhibitor or the combination of the SETBP1 inhibitor and the XPO1 inhibitor is present in a pharmaceutical composition comprising one or more excipients.
41. A method of inducing embryonic or fetal hemoglobin expression in a cell, wherein the cell is a hematopoietic progenitor cell, an erythroid progenitor cell, or an erythroid cell, comprising contacting the cell with a SETBP1 inhibitor, an XPO1 inhibitor, or a combination of a SETBP1 inhibitor and an XPO1 inhibitor.
42. The method of claim 41, wherein the SETBP1 inhibitor is a compound of Formula (1):
Figure US20220249486A1-20220811-C00023
wherein:
R1 is —OR8;
R2, R3, and R6 are H;
R4 and R5 are independently C1-4 alkyl;
R7 is —NR10R10 or a piperidinyl group;
L is —(CR9R9)n—;
X is an organic or inorganic anion;
R8 is H or C1-3 alkyl;
each R9 is independently H or C1-3 alkyl;
each R10 is independently H or C1-3 alkyl; and
n is 1, 2, 3 or 4.
43. The method of claim 42, wherein X is selected from the group consisting of F, Cl, Br, and I.
44. The method of claim 42, wherein
R1 is —OR8;
R2, R3, R6, R8, and R9 are H;
R4 and R5 are —CH3;
R7 is a piperidinyl group;
L is —(CR9R9)n—;
X is selected from the group consisting of F, Cl, Br, and I; and
n is 1, 2, 3 or 4.
45. The method of claim 42, wherein
R1 is —OR8;
R2, R3, R6, R8 and R9 are H;
R4 and R5 are —CH3;
R7 is —NR10R10;
each R10 is independently C1-3 alkyl;
L is —(CR9R9)n—;
X is selected from the group consisting of F, Cl, Br, and I; and
n is 1, 2, 3 or 4.
46. The method of claim 42, wherein the compound of Formula (1) is a compound of Formula (1A)
Figure US20220249486A1-20220811-C00024
wherein X is selected from the group consisting of F, Cl, Br, and I.
47. The method of claim 42, wherein the compound of Formula (1) is Compound (1AA)
Figure US20220249486A1-20220811-C00025
48. The method of claim 42, wherein the compound of Formula (1) is a compound of Formula (1B)
Figure US20220249486A1-20220811-C00026
wherein X is selected from the group consisting of F, Cl, Br, and I.
49. The method of claim 42, wherein the compound of Formula (1) is Compound (1BB)
Figure US20220249486A1-20220811-C00027
50. The method of claim 41, wherein the SETBP1 inhibitor is a compound of Formula (2A)
Figure US20220249486A1-20220811-C00028
wherein HA is a proton donor.
51. The method of claim 50, wherein HA is selected from the group consisting of hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, and nitric acid.
52. The method of any one of claims 50-51, wherein the compound of Formula (2A) is Compound (2AA)
Figure US20220249486A1-20220811-C00029
53. The method of claim 41, wherein the SETBP1 inhibitor is an shRNA or an siRNA.
54. The method of claim 53, wherein the shRNA or the siRNA has a sequence that is at least 95% identical to a nucleic acid sequence selected from the group consisting of SEQ ID NO: 1 and SEQ ID NO: 2.
55. The method of claim 41, wherein the SETBP1 inhibitor is a gene editing system, and wherein the gene editing system is selected from the group consisting of CRISPR/Cas9, CRISPR/Cas13, a zinc finger nuclease system, a TALEN system, and a meganuclease system.
56. The method of claim 41, wherein the XPO1 inhibitor is selected from the group consisting of KPT-185, KPT-276, KPT-330, KPT-335 and KPT-8602.
57. The method of claim 41, wherein the XPO1 inhibitor is an shRNA or an siRNA.
58. The method of claim 41, wherein the XPO1 inhibitor is a gene editing system, and wherein the gene editing system is selected from the group consisting of CRISPR/Cas9, CRISPR/Cas13, a zinc finger nuclease system, a TALEN system, and a meganuclease system.
US17/616,158 2019-06-06 2020-06-04 Setbp1 and xpo1 inhibitors for the treatment of sickle cell disease and beta-thalassemia Pending US20220249486A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/616,158 US20220249486A1 (en) 2019-06-06 2020-06-04 Setbp1 and xpo1 inhibitors for the treatment of sickle cell disease and beta-thalassemia

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201962858103P 2019-06-06 2019-06-06
US17/616,158 US20220249486A1 (en) 2019-06-06 2020-06-04 Setbp1 and xpo1 inhibitors for the treatment of sickle cell disease and beta-thalassemia
PCT/US2020/036151 WO2020247654A1 (en) 2019-06-06 2020-06-04 Setbp1 and xpo1 inhibitors for the treatment of sickle cell disease and beta-thalassemia

Publications (1)

Publication Number Publication Date
US20220249486A1 true US20220249486A1 (en) 2022-08-11

Family

ID=73652619

Family Applications (1)

Application Number Title Priority Date Filing Date
US17/616,158 Pending US20220249486A1 (en) 2019-06-06 2020-06-04 Setbp1 and xpo1 inhibitors for the treatment of sickle cell disease and beta-thalassemia

Country Status (2)

Country Link
US (1) US20220249486A1 (en)
WO (1) WO2020247654A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20210205306A1 (en) 2018-05-24 2021-07-08 The Henry M. Jackson Foundation For The Advancement Of Military Medicine, Inc. Setbp1 inhibitors for the treatment of myeloid neoplasms and solid tumors

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014204723A1 (en) * 2013-06-17 2014-12-24 The Broad Institute Inc. Oncogenic models based on delivery and use of the crispr-cas systems, vectors and compositions
US20160237455A1 (en) * 2013-09-27 2016-08-18 Editas Medicine, Inc. Crispr-related methods and compositions
ES2739637T3 (en) * 2013-11-28 2020-02-03 Inst Nat Sante Rech Med Pharmaceutical methods and compositions for the treatment of beta-thalassemias
US11179412B2 (en) * 2017-12-04 2021-11-23 University of Pittsburgh—of the Commonwealth System of Higher Education Methods of treating conditions involving elevated inflammatory response
US20210205306A1 (en) * 2018-05-24 2021-07-08 The Henry M. Jackson Foundation For The Advancement Of Military Medicine, Inc. Setbp1 inhibitors for the treatment of myeloid neoplasms and solid tumors

Also Published As

Publication number Publication date
WO2020247654A1 (en) 2020-12-10

Similar Documents

Publication Publication Date Title
JP5478262B2 (en) Anticancer drug
RU2316326C2 (en) Method and composition for cancer disease treatment, tosylate and pharmaceutically acceptable salts of n-(4-chloro-3-(trifluoromethyl)phenyl)-n&#39;-(4-(2-(n-methylcarbamoyl)-4-pyridyloxy)phenyl)urea
US6365583B1 (en) Methods to enhance white blood cell count
Zhang et al. Supramolecular therapeutics to treat the side effects induced by a depolarizing neuromuscular blocking agent
CN107406438A (en) The inhibitor of bromine domain
KR20140107174A (en) Treatment of breast cancer
US20200197392A1 (en) Compositions and methods for treating tuberous sclerosis complex
AU697742B2 (en) Treatment of cytokine growth factor caused disorders
US20220249486A1 (en) Setbp1 and xpo1 inhibitors for the treatment of sickle cell disease and beta-thalassemia
Miller et al. Trpm2 enhances physiological bioenergetics and protects against pathological oxidative cardiac injury: Role of Pyk2 phosphorylation
US20210205306A1 (en) Setbp1 inhibitors for the treatment of myeloid neoplasms and solid tumors
US20130324520A1 (en) Rxrg modulators for the treatment of cancer
Fowler et al. Targeting the canonical nuclear factor-κB pathway with a high-potency IKK2 inhibitor improves outcomes in a mouse model of idiopathic pneumonia syndrome
US10266490B2 (en) Radioprotector compounds
JP2015510945A (en) Methods of treating cancer using aurora kinase inhibitors
CN113662942B (en) Pharmaceutical composition and application thereof in SMO (small molecule allowed) mutant medulloblastoma
CN114469940B (en) Application of small molecule compound AQ-390 in preparing medicine and inhibitor for resisting cell apoptosis
JPWO2020004404A1 (en) IL-1β inhibitor
EP3638690A1 (en) Methods to treat gliomas using a stat3 inhibitor
WO2004035056A1 (en) Use of skca channel blocking drugs for combating parkinson&#39;s disease
CN113456643B (en) Pharmaceutical composition containing plinabulin and application thereof
WO2022127788A1 (en) Application of lenvatinib and aurora-a kinase inhibitor in preparing cancer-inhibiting drugs
US20230255961A1 (en) Rotomeric Isomers of 4-Alkyl-5-Heteroaryl-3H-1,2-Dithiole-3-Thiones
CN116271057A (en) Combined pharmaceutical composition for preventing or treating follicular lymphoma and application thereof
CN117883455A (en) Combined pharmaceutical composition for preventing or treating MLL rearrangement acute myelogenous leukemia and application thereof

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION